Language selection

Search

Patent 2897828 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 2897828
(54) English Title: METHODS FOR IDENTIFYING, DIAGNOSING, AND PREDICTING SURVIVAL OF LYMPHOMAS
(54) French Title: PROCEDES POUR IDENTIFIER ET DIAGNOSTIQUER LES LYMPHOMES ET PREDIRE LA DUREE DE LEUR SURVIE
Status: Granted and Issued
Bibliographic Data
(51) International Patent Classification (IPC):
  • C12Q 01/6809 (2018.01)
  • C12Q 01/68 (2018.01)
  • C12Q 01/6813 (2018.01)
  • G16B 05/00 (2019.01)
  • G16B 25/10 (2019.01)
(72) Inventors :
  • STAUDT, LOUIS M. (United States of America)
  • WRIGHT, GEORGE (United States of America)
  • DAVE, SANDEEP (United States of America)
  • TAN, BRUCE (United States of America)
  • POWELL, JOHN I. (United States of America)
  • WILSON, WYNDHAM (United States of America)
  • JAFFE, ELAINE (United States of America)
  • CHAN, WING C. (United States of America)
  • GREINER, TIMOTHY C. (United States of America)
  • WEISENBURGER, DENNIS (United States of America)
  • ARMITAGE, JAMES (United States of America)
  • FU, KAI (United States of America)
  • FISHER, RICHARD I. (United States of America)
  • RIMSZA, LISA (United States of America)
  • MILLER, THOMAS (United States of America)
  • GROGAN, THOMAS (United States of America)
  • GUERRI, ELIAS CAMPO (Spain)
  • BEA, SILVIA M. (Spain)
  • SALAVERRIA, ITZIAR (Spain)
  • LOPEZ-GUILLERMO, ARMANDO (Spain)
  • MONTSERRAT COSTA, EMILIO (Spain)
  • MORENO, VICTOR (Spain)
  • ZETTL, ANDREAS (Germany)
  • OTT, GERMAN (Germany)
  • MULLER-HERMELINK, HANS-KONRAD (Germany)
  • ROSENWALD, ANDREAS (Germany)
  • VOSE, JULIE (United States of America)
  • SMELAND, ERLEND (Norway)
  • KVALOY, STEIN (Norway)
  • HOLTE, HARALD (Norway)
  • DELABIE, JAN (Norway)
  • LISTER, ANDREW (United Kingdom)
  • CONNORS, JOSEPH (Canada)
  • GASCOYNE, RANDY (Canada)
(73) Owners :
  • THE UNITED STATES OF AMERICA, REPRESENTED BY THE SECRETARY, DEPARTMENT O
  • THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMEN
  • BOARD OF REGENTS OF THE UNIVERSITY OF NEBRASKA
  • UNIVERSITY OF ROCHESTER
  • QUEEN MARY AND WESTFIELD COLLEGE, UNIVERSITY OF LONDON
  • BRITISH COLUMBIA CANCER AGENCY BRANCH
  • OSLO UNIVERSITY HOSPITAL HF
  • ARIZONA BOARD OF REGENTS ON BEHALF OF THE UNIVERSITY OF ARIZONA
  • INSTITUT CATALA D'ONCOLOGIA
  • UNIVERSITAT DE BARCELONA
  • FUNDACIO CLINIC
  • HOSPITAL CLINIC
  • JULIUS-MAXIMILIANS-UNIVERSITY OF WURZBURG
(71) Applicants :
  • THE UNITED STATES OF AMERICA, REPRESENTED BY THE SECRETARY, DEPARTMENT O (United States of America)
  • THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMEN (United States of America)
  • BOARD OF REGENTS OF THE UNIVERSITY OF NEBRASKA (United States of America)
  • UNIVERSITY OF ROCHESTER (United States of America)
  • QUEEN MARY AND WESTFIELD COLLEGE, UNIVERSITY OF LONDON (United Kingdom)
  • BRITISH COLUMBIA CANCER AGENCY BRANCH (Canada)
  • OSLO UNIVERSITY HOSPITAL HF (Norway)
  • ARIZONA BOARD OF REGENTS ON BEHALF OF THE UNIVERSITY OF ARIZONA (United States of America)
  • INSTITUT CATALA D'ONCOLOGIA (Spain)
  • UNIVERSITAT DE BARCELONA (Spain)
  • FUNDACIO CLINIC (Spain)
  • HOSPITAL CLINIC (Spain)
  • JULIUS-MAXIMILIANS-UNIVERSITY OF WURZBURG (Germany)
(74) Agent: SMART & BIGGAR LP
(74) Associate agent:
(45) Issued: 2022-06-21
(22) Filed Date: 2007-07-25
(41) Open to Public Inspection: 2008-01-31
Examination requested: 2015-07-21
Availability of licence: N/A
Dedicated to the Public: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): No

(30) Application Priority Data:
Application No. Country/Territory Date
11/493,387 (United States of America) 2006-07-25

Abstracts

English Abstract

Gene expression data provides a basis for more accurate identification and diagnosis of lymphoproliferative disorders. In addition, gene expression data can be used to develop more accurate predictors of survival. The present invention discloses methods for identifying, diagnosing, and predicting survival in a lymphoma or lymphoproliferative disorder on the basis of gene expression patterns. The invention discloses a novel microarray, the Lymph Dx microarray, for obtaining gene expression data from a lymphoma sample. The invention also discloses a variety of methods for utilizing lymphoma gene expression data to determine the identity of a particular lymphoma and to predict survival in a subject diagnosed with a particular lymphoma. This information will be useful in developing the therapeutic approach to be used with a particular subject.


French Abstract

Des données dexpression génique fournissent une base pour une identification et un diagnostic plus précis des troubles lymphoprolifératifs. De plus, lesdites données peuvent être utilisées pour développer des prédicteurs de survie plus précis. La présente invention concerne des procédés pour identifier et diagnostiquer des lymphomes, et prédire la survie dun lymphome ou dun trouble lymphoprolifératif à partir de motifs dexpression génique. Linvention concerne un nouveau microréseau, le microréseau Lymph Dx, pour obtenir des données dexpression génique à partir dun échantillon de lymphome. Linvention concerne également une diversité de procédés pour utiliser les données dexpression génique de lymphome pour déterminer lidentité dun lymphome particulier et pour prédire la survie dun sujet pour lequel un lymphome particulier a été diagnostiqué. Ces informations seront utiles dans le développement de lapproche thérapeutique à utiliser pour un sujet particulier.

Claims

Note: Claims are shown in the official language in which they were submitted.


WE CLAIM:
1. A method for predicting survival in a diffuse large B cell lymphoma
(DLBCL) subject,
wherein the method comprises:
a) selecting a biopsy sample from a DLBCL subject,
b) assaying the sample for gene expression levels for a set of genes
expressed in the
sample,
c) determining an average gene expression level of genes set forth below
<IMG>
200

<IMG>
for the proliferation gene expression signature to obtain a proliferation gene
expression signature
value;
d) determining an average gene expression level of genes set forth in the
table of (c)
for the germinal center B cell gene expression signature to obtain a germinal
center B cell gene expression signature value;
e) determining an average gene expression level of genes set forth in the
table of (c)
for the MHC class II gene expression signature to obtain an MHC class II gene
expression signature value;
determining an average gene expression level of genes set forth in the table
of (c)
for the lymph node gene expression signature to obtain a lymph node gene
expression signature value;
measuring the expression level of B/1696 to obtain a B11696 expression value;
h) calculating a survival predictor score using an equation:
[0.241*(proliferation
gene expression signature value)] + [0.310*(B/1696 expression value)] ¨
[0.290*(germinal center B cell gene expression signature value)] ¨ [0.311*(MHC
class II gene expression signature value)] ¨ [0.249*(lymph node gene
expression
201

signature value)], wherein a higher survival predictor score is associated
with
worse survival prognosis.
2. The method of claim 1, wherein the method further comprises assaying the
sample for a
gain or amplification in the 3p11-p12 region of chromosome 3, wherein a gain
or
amplification in the 3p11-p12 region of chromosome 3 indicates that the
survival
predictor score should be increased.
3. The method of claim 2, wherein the assaying for a gain or amplification
in the 3p11-p12
region of chromosome 3 comprises performing comparative genomic hybridization.
4. The method of claim 2, wherein the assaying for a gain or amplification
in the 3p11-p12
region of chromosome 3 comprises performing cytogenetic analysis.
5. The method of claim 2, wherein the assaying for a gain or amplification
in the 3p11-p12
region of chromosome 3 comprises performing polymerase chain reaction (PCR).
6. The method of claim 5, wherein the PCR is real-time quantitative PCR.
202

Description

Note: Descriptions are shown in the official language in which they were submitted.


METHODS FOR IDENTIFYING, DIAGNOSING, AND PREDICTING SURVIVAL OF
LYMPHOMAS
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH AND
DEVELOPMENT
[0001] This invention iwas made with U.S. Government support under project
number ZIA BC 011006 by the National Institutes of Health, national Cancer
Institute. National Cancer Institute. The U.S. Government has certain rights
in the
invention.
REFERENCE TO TABLES SUBMITTED ON COMPACT DISC
[0002] Tables 2-1723 and 1725-2358 are contained on 22 CD-ROMs submitted
herewith. The CD-ROMs submitted herewith are numbered 1-21 of 22. The name,
size, and date of creation for each file is presented in the file entitled
"Table_of_contents.txt," located on CD number 21 of 22. The name of each file
incorporates the number of the corresponding table.
REFERENCE TO COMPUTER PROGRAM LISTING APPENDIX SUBMITTED ON
COMPACT DISC
[0003] A computer program listing appendix is contained on one CD-ROM
submitted herewith, which is numbered 22 of 22. The computer program listing
appendix contains files related to the implementation of an algorithm for
determining
lymphoma type. The name, size, and date of creation for each file in the
computer
program listing appendix is presented in the file entitled
"Table_of_contents.txt,"
located on CD-ROM 22.
BACKGROUND
[0004] A variety of systems for identifying and classifying lymphomas have
been
proposed over the last 20 years. In the 1980's, the Working Formulation was
introduced as a method of classifying lymphomas based on morphological and
1
CA 2897828 2019-12-20

clinical characteristics. In the 1990's, the Revised European-American
Lymphoma
(REAL) system was introduced in an attempt to take into account
immunophenotypic
and genetic characteristics in classifying lymphomas (Harris 1994). The most
recent
standard, set forth by the World Health Organization (WHO), attempts to build
on
these previous systems (Jaffe 2001). The WHO classification of lymphomas is
based on several factors, including tumor morphology, immunophenotype,
recurrent
genetic abnormalities, and clinical features. Table 1, below, contains a list
of the B
and T cell neoplasms that have been recognized by the WHO classification. Each
malignancy is listed according to its WHO classification nomenclature,
followed by a
WHO classification number.
Table 1
B-cell neoplasms
Category Name WHO ID #
Precursor B-cell neoplasms Precursor B-cell lymphoblastic 9835/3
leukemia
Precursor B-cell lymphoblastic 9728/3
lymphoma
Mature B-cell neoplasms Chronic lymphocytic leukemia 9823/3
Small lymphocytic lymphoma 9670/3
B-cell prolymphocytic leukemia 9833/3
Lymphoplasmacytic lymphoma 9671/3
Splenic marginal zone 9689/3
lymphoma
Hairy cell leukemia 9940/3
Plasma cell myeloma 9732/3
Solitary plasmacytoma of bone 9731/3
Extraosseous plasmacytonna 9734/3
Extranodal marginal zone B-cell 9699/3
lymphoma of mucosa-associated
lymphoid tissue (MALT lymphoma)
Nodal marginal zone B-cell 9699/3
lymphoma
Follicular lymphoma (Grade 1, 2, 9690/3
3a, 3b)
Mantle cell lymphoma 9673/3
Diffuse large B-cell lymphoma 9680/3
Mediastinal (thymic) large B-cell 9679/3
lymphoma
Intrayascular large B-cell 9680/3
lymphoma
Primary effusion lymphoma 9678/3
Burkitt lymphoma 9687/3
Burkitt leukemia 9826/3
2
CA 2 8 9 7 8 2 8 2 0 1 9-1 2-2 0

B-cell proliferations of uncertain Lymphomatoid
granulomatosis 9766/1
malignant potential
Post-transplant lymphoproliferative 9970/1
disorder, polymorphic
1-cell and NK-cell neoplasms
Precursor T-cell and NK-cell Precursor T lymphoblastic 9837/3
neoplasms leukemia
Precursor T lymphoblastic 9729/3
lymphoma
Blastic NK-cell lymphoma 9727/3
Mature T-cell and NK-cell T-cell prolymphocytic leukemia 9834/3
neoplasms
T-cell large granular lymphocytic 9831/3
leukemia
Aggressive NK-cell leukemia 9948/3
Adult 1-cell leukemia/lymphoma 9827/3
Extranodal NK-/T-cell lymphoma, 9719/3
nasal type
Enteropathy-type 1-cell lymphoma 9717/3
Hepatosplenic T-cell lymphoma 9716/3
Subcutaneous panniculitis-like T- 9708/3
cell lymphoma
Mycosis fungoides 9700/3
Sezary syndrome (9701/3) 9701/3
Primary cutaneous anaplastic large 9718/3
cell lymphoma (C-ALCL)
Peripheral T-cell lymphoma, 9702/3
unspecified
Angioimmunoblastic T-cell 9705/3
lymphoma
Anaplastic large cell lymphoma 9714/3
T-cell proliferation of uncertain Lymphomatoid papulosis
9718/3
malignant potential
Hodgkin lymphoma Nodular lymphocyte predominant 9659/3
Hodgkin lymphoma
Classical Hodgkin lymphoma 9650/3
Classical Hodgkin lymphoma, 9663/3
nodular sclerosis
Classical Hodgkin lymphoma, 9651/3
lymphocyte-rich
Classical Hodgkin lymphoma, 9652/3
mixed cellularity
Classical Hodgkin lymphoma, 9653/3
lymphocyte depleted
Other diagnoses that have not been given WHO diagnostic numbers include HIV-
associated lymphoma, germinal center B cell-like subtype of diffuse large B
cell
lymphoma, activated B cell-like subtype of diffuse large B-cell lymphoma,
follicular
hyperplasia (non-malignant), and infectious mononucleosis (non-malignant).
3
CA 2 8 9 7 8 2 8 2 0 1 9-1 2-2 0

[0005] Although the WHO classification has proven useful in patient management
and treatment, patients assigned to the same WHO diagnostic category often
have
noticeably different clinical outcomes. In many cases, these different
outcomes
appear to be due to molecular differences between tumors that cannot be
readily
observed by analyzing tumor morphology. More precise methods are needed for
identifying and classifying lymphomas based on their molecular
characteristics.
SUMMARY
[0006] In certain embodiments, a composition is provided comprising the set of
probes listed in Table 2, which is contained in the file entitled
"Table_0002_LymphDx_Probe_List.txt." Preferably, this composition comprises a
microarray.
[0007] In certain embodiments, a method is provided for generating a survival
predictor for a particular lymphoma type. In this method, one or more biopsy
samples that have been diagnosed as belonging to a particular lymphoma type
are
obtained. Gene expression data is obtained for these samples, and genes with
expression patterns associated with longer or shorter survival are identified.
Hierarchical clustering is performed to group these genes into gene expression
signatures, and the expression of all genes within each signature are averaged
to
obtain a gene expression signature value for each signature. These gene
expression signature values are then used to generate a multivariate survival
predictor.
[0008] In certain embodiments, a method is provided for predicting survival in
a
follicular lymphoma (FL) subject. In this method, a biopsy sample is obtained
from
the subject and gene expression data is obtained from the biopsy sample. The
expression level of those genes belonging to an immune response-1 or immune
4
CA 2897828 2019-12-20

response-2 gene expression signature are averaged to generate gene expression
signature values for each signature. A survival predictor score is then
calculated
using an equation: [2.71*(immune response-2 gene expression signature value)]
¨
[2.36*(immune response-1 gene expression signature value)]. A higher survival
predictor score is associated with a less favorable outcome. In one
embodiment,
the gene expression data used in this method is obtained using a microarray.
[0009] In certain embodiments, another method is provided for predicting
survival in
a follicular lymphoma (FL) subject. In this method, a biopsy sample is
obtained from
the subject and gene expression data is obtained from the biopsy sample. The
expression level of those genes belonging to a B cell differentiation, T-cell,
or
macrophage gene expression signature are averaged to generate gene expression
signature values for each signature. A survival predictor score is then
calculated
using an equation: [2.053*(macrophage gene expression signature value)] ¨
[2.344*(T-cell gene expression signature value)] ¨ [0.729*(B-cell gene
expression
signature value)]. A higher survival predictor score is associated with a less
favorable outcome. In one embodiment, the gene expression data used in this
method is obtained using a microarray.
[0010] In certain embodiments, yet another method is provided for predicting
survival in a follicular lymphoma (FL) subject. In this method, a biopsy
sample is
obtained from the subject and gene expression data is obtained from the biopsy
sample. The expression level of those genes belonging to a macrophage, 1-cell,
or
B-cell differentiation gene expression signature are averaged to generate gene
expression signature values for each signature. A survival predictor score is
then
calculated using an equation: [1.51*(macrophage gene expression signature
value)]
¨ [2.11*(T-cell gene expression signature value)] ¨ [0.505*(B-cell
differentiation gene
CA 2897828 2019-12-20

expression signature value)]. A higher survival predictor score is associated
with a
less favorable outcome. In one embodiment, the gene expression data used in
this
method is obtained using a microarray.
[0011] In certain embodiments, a method is provided for predicting survival in
a
diffuse large B cell lymphoma (DLBCL) subject. In this method, a biopsy sample
is
obtained from the subject and gene expression data is obtained from the biopsy
sample. The expression level of those genes belonging to an ABC DLBCL high,
lymph node, or MHC class II gene expression signature are averaged to generate
gene expression signature values for each signature. A survival predictor
score is
then calculated using an equation: [0.586*(ABC DLBCL high gene expression
signature value)] ¨ [0.468*(lymph node gene expression signature value)] ¨
[0.336*(MHC class II gene expression signature value)]. A higher survival
predictor
score is associated with a less favorable outcome. In one embodiment, the gene
expression data used in this method is obtained using a microarray.
[0012] In certain embodiments, another method is provided for predicting
survival in
a diffuse large B cell lymphoma (DLBCL) subject. In this method, a biopsy
sample is
obtained from the subject and gene expression data is obtained from the biopsy
sample. The expression level of those genes belonging to a lymph node,
germinal B
cell, proliferation, or MHC class II gene expression signature are averaged to
generate gene expression signature values for each signature. A survival
predictor
score is then calculated using an equation: [-0.4337*(lymph node gene
expression
signature)] + [0.09*(proliferation gene expression signature)] ¨
[0.4144*(germinal
center B-cell gene expression signature)] ¨ [0.2006*(MHC class II gene
expression
signature)]. A higher survival predictor score is associated with a less
favorable
6
CA 2897828 2019-12-20

outcome. In one embodiment, the gene expression data used in this method is
obtained using a microarray.
[0013] In certain embodiments, yet another method is provided for predicting
survival in a diffuse large B cell lymphoma (DLBCL) subject. In this method, a
biopsy sample is obtained from the subject and gene expression data is
obtained
from the biopsy sample. The expression level of those genes belonging to a
lymph
node, germinal B cell, or MHC class II gene expression signature are averaged
to
generate gene expression signature values for each signature. A survival
predictor
score is then calculated using an equation: [-0.32*(lymph node gene expression
signature)] ¨ [0.176*(germinal B cell gene expression signature)] ¨
[0.206*(MHC
class ll gene expression signature)]. A higher survival predictor score is
associated
with a less favorable outcome. In one embodiment, the gene expression data
used
in this method is obtained using a microarray. In another embodiment, the gene
expression data is obtained using RT-PCR.
[0014] In certain embodiments, a method is provided for refining a method for
predicting survival in a diffuse large B cell lymphoma (DLBCL). In this
method, gains
or amplifications of the 3p11-p12 region in chromosome 3 are identified,
wherein
gains or amplifications of this region are associated with a less favorable
outcome.
This information can be integrated into an existing method for predicting
DLBCL
survival. For example, a survival predictor score may be calculated using an
equation such as:
Survival predictor score = [0.241*(proliferation gene expression
signature value)] + [0.310*(BMP6 expression value)] ¨
[0.290*(germinal center B cell gene expression signature value)] ¨
7
CA 2897828 2019-12-20

[0.311*(MHC class II gene expression signature value)] ¨
[0.249*(lymph node gene expression signature value)],
wherein a higher survival predictor score is associated with a less favorable
outcome. The DLBCL sample may then be assayed for gains or amplifications in
the 3p11-p12 using any available method, including for example CGH. The
identification of gains or amplifications in this region indicate a decrease
in survival.
This information can be used to adjust the survival predictor score
accordingly.
Alternatively, identification of a gain or amplification of 3p11-p12 may be
used as a
stand-alone indicator of less favorable outcome for a DLBCL patient in the
absence
of additional gene expression data.
[0015] In certain embodiments, a method is provided for predicting survival in
a
mantle cell lymphoma (MCL) subject. In this method, a biopsy sample is
obtained
from the subject and gene expression data is obtained from the biopsy sample.
The
expression level of those genes belonging to a proliferation gene expression
signature are averaged to generate a gene expression signature value. A
survival
predictor score is then calculated using an equation: [1.66*(proliferation
gene
expression signature value)]. A higher survival predictor score is associated
with a
less favorable outcome. In one embodiment, the gene expression data used in
this
method is obtained using a microarray.
[0016] In certain embodiments, a method is provided for determining the
probability
that a sample X belongs to a first lymphoma type or a second lymphoma type. In
this method, a set of genes is identified that is differentially expressed
between the
two lymphoma types in question, and a set of scale factors representing the
difference in expression between the lymphoma types for each of these genes
are
calculated. A series of linear predictor scores are generated for samples
belonging
8
CA 2897828 2019-12-20

to either of the two lymphoma types based on expression of these genes. Gene
expression data is then obtained for sample X, and a linear predictor score is
calculated for this sample. The probability that sample X belongs to the first
lymphoma type is calculated using an equation that incorporates the linear
predictor
score of sample X and the mean and variance of the linear predictor scores for
the
known samples of either lymphoma type.
[0017] In certain embodiments, a method is provided for determining the
lymphoma
type of a sample X. In this method, a set of genes is identified that is
differentially
expressed between a first lymphoma type and a second lymphoma type, and a set
of scale factors representing the difference in expression of each of these
genes
between the two lymphoma types are calculated. A series of linear predictor
scores
are generated for samples belonging to either of the two lymphoma types based
on
expression of these genes. Gene expression data is then obtained for sample X,
and a linear predictor score is calculated for this sample. The probability
that
sample X belongs to the first lymphoma type is calculated using an equation
that
incorporates the linear predictor score of sample X and the mean and variance
of
the linear predictor scores for the known samples of either lymphoma type.
This
entire process is then repeated with various lymphoma types being substituted
for
the first lymphoma type, the second lymphoma type, or both.
[0018] In certain embodiments, a method is provided for determining the
lymphoma
type of a sample X. In this method, a series of lymphoma type pairs are
created,
with each pair consisting of a first lymphoma type and a second lymphoma type.
For
each type pair, gene expression data is obtained for a set of genes, and a
series of
scale factors representing the difference in expression of each of these genes
between the two lymphoma types are calculated. A subset of z genes with the
9
CA 2897828 2019-12-20

largest scale factors are identified, and a series of linear predictor scores
are
generated for samples belonging to either of the two lymphoma types. Linear
predictor scores are calculated for anywhere from 1 to z of these genes. The
number of genes from 1 to z that results in the largest difference in linear
predictor
scores between the two lymphoma types is selected, and gene expression data
for
these genes is obtained for sample X. A linear predictor score is generated
for
sample X, and the probability that the sample belongs to the first lymphoma
type is
calculated using an equation that incorporates the linear predictor score for
sample
X and the mean and variance of the linear predictor scores for the known
samples of
either lymphoma type.
[0019] In certain embodiments, another method is provided for determining the
lymphoma type of a sample X. In this method, a series of lymphoma type pairs
are
created, with each pair consisting of a first lymphoma type and a second
lymphoma
type. For each type pair, gene expression data is obtained for a set of genes,
and a
series of scale factors representing the difference in expression of each of
these
genes between the two lymphoma types are calculated. The set of genes is
divided
into gene-list categories indicating correlation with a gene expression
signature.
Within each gene-list category, a subset of z genes with the largest scale
factors are
identified, and a series of linear predictor scores are generated for samples
belonging to either of the two lymphoma types. Linear predictor scores are
calculated for anywhere from 1 to z of these genes. The number of genes from 1
to
z that results in the largest difference in linear predictor scores between
the two
lymphoma types is selected, and gene expression data for these genes is
obtained
for sample X. A linear predictor score is generated for sample X, and the
probability
q that the sample belongs to the first lymphoma type is calculated using an
equation
CA 2897828 2019-12-20

that incorporates the linear predictor score for sample X and the mean and
variance
of the linear predictor scores for the known samples of either lymphoma type.
A
high probability q indicates that sample X belongs to the first lymphoma type,
a low
probability q indicates that sample X belongs to the second lymphoma type, and
a
middle probability q indicates that sample X belongs to neither lymphoma type.
The
cut-off point between high, middle, and low probability values is determined
by
ranking samples of known lymphoma type according to their probability values,
then
analyzing every possible cut-off point between adjacent samples by: 3.99*[(%
of first
lymphoma type misidentified as second lymphoma type) + (% of second lymphoma
type misidentified as a first lymphoma type)] + [(Y of first lymphoma type
identified
as belonging to neither lymphoma type) + (% of second lymphoma type identified
as
belonging to neither lymphoma type)]. The final cut-off points are those that
minimize the value of this equation.
[0020] In certain embodiments, a method is provided for classifying a sample
as
BL. In this method, a series of lymphoma type pairs are created, with each
pair
consisting of a first lymphoma type (BL) and a second lymphoma type. In
certain
embodiments, the second lymphoma type is DLBCL, and in certain of these
embodiments, the DLBCL is ABC, GCB, or PMBL. For each type pair, gene
expression data is obtained for a set of genes, and a series of scale factors
representing the difference in expression of each of these genes between the
two
lymphoma types are calculated. The set of genes is divided into two subsets.
The
first subset includes c-myc and c-myc target genes, while the second subset
includes z genes from the gene set that exhibit the largest scale factors and
do not
fall into the first subset. In certain embodiments, z is 100. A first and
second series
of linear predictor scores are generated for samples belonging to either of
the two
11
CA 2897828 2019-12-20

lymphoma types, with the first series based on expression of the first gene
subset
and the second series based on expression of the second gene subset.
Expression
of the first and second gene subsets is measured for sample X, and a first and
second linear predictor score is generated for sample X based on the
expression of
the first and second gene subsets, respectively. Two probabilities, qi and q2,
are
calculated using equations that incorporate the first and second linear
predictor
scores for sample X, respectively, and the mean and variance of the linear
predictor
scores for the known samples of either lymphoma type. Sample X is classified
as
BL if both qi and q2 are greater than 90%.
[0021] In certain embodiments, a method is provided for identifying a lymphoma
sample as cyclin Dl-negative MCL. In this method, a candidate sample X is
identified based on a lack of cyclin D1 expression. A series of lymphoma type
pairs
are created, with each pair consisting of MCL and a second lymphoma type. For
each type pair, gene expression data is obtained for a set of genes G, and a
series
of scale factors representing the difference in expression of each of these
genes
between the two lymphoma types are calculated. A subset of genes g with the
largest scale factors are identified, and a series of linear predictor scores
are
generated for samples belonging to either of the two lymphoma types. Gene
expression data for the subset of genes g is obtained for sample X. A linear
predictor score is generated for sample X, and the probability that the sample
belongs to the first lymphoma type is calculated using an equation that
incorporates
the linear predictor score for sample X and the mean and variance of the
linear
predictor scores for the known samples of either lymphoma type. A probability
greater than 90% indicates that the sample X is cyclin Dl-negative MCL.
12
CA 2897828 2019-12-20

BRIEF DESCRIPTION OF THE DRAWINGS
[0022] Figure 1: Method for identifying lymphoma type. Flow chart depicts a
general method for identifying lymphoma type using gene expression data.
[0023] Figure 2: Survival signature analysis. Flow chart depicts method for
developing a lymphoma survival predictor based on gene expression patterns.
[0024] Figure 3: FL survival data. Survival data for 191 subjects diagnosed
with FL.
Median age at diagnosis was 51 years (ranging from 23 to 81 years), and the
subjects had a median follow-up of 6.6 years (8.1 years for survivors, with a
range of
<1 to 28.2 years).
[0025] Figure 4: Hierarchical clustering of survival associated genes in FL
samples.
Each column represents a single FL sample, while each row represents a single
gene. Relative gene expression is depicted according to the scale at the
bottom of
the figure. The dendrogram to the left indicates the degree to which the
expression
pattern of each gene is correlated with that of the other genes. The bars
indicate
sets of coordinately regulated genes defined as gene expression signatures.
Genes
comprising the immune response-1 and immune response-2 gene expression
signature are listed on the right.
[0026] Figure 5: Kaplan-Meier plot of survival in FL samples based on survival
predictor scores. 191 FL samples were divided into quartiles based on their
survival
predictor scores. The survival predictor scores were calculated by:
[(2.71*immune
response-2 gene expression signature value)] ¨ [(2.36 x immune response-1 gene
expression signature value)].
[0027] Figure 6: Kaplan-Meier plot of survival in FL samples based on IPI
score. 96
FL samples were divided into three groups based on their IPI scores.
13
CA 2897828 2019-12-20

[0028] Figure 7: Kaplan-Meier plot of survival in FL samples with low or high
risk IPI
scores based on survival predictor scores. 96 FL samples with low risk (left
panel)
or intermediate risk (right panel) IPI scores were divided into quartiles
based on their
survival predictor scores. The survival predictor scores were calculated by:
[(2.71*immune response-2 gene expression signature value)] ¨ [(2.36 x immune
response-1 gene expression signature value)].
[0029] Figure 8: Kaplan-Meier plot of survival in FL samples based on survival
predictor scores. 191 FL samples were divided into quartiles based on their
survival
predictor scores. The survival predictor scores were calculated by:
[2.053*(macrophage gene expression signature value)] ¨ [2.344*(T-cell gene
expression signature value)] ¨ [0.729*(B-cell differentiation gene expression
signature value)].
[0030] Figure 9: Kaplan-Meier plot of survival in FL samples based on survival
predictor scores. 191 FL samples were divided into quartiles based on their
survival
predictor scores. The survival predictor scores were calculated by:
[1.51*(macrophage gene expression signature value)] ¨ [2.11*(T-cell gene
expression signature value)] ¨ [0.505*(B-cell differentiation gene expression
signature value)].
[0031] Figure 10: Kaplan-Meier plot of survival in DLBCL samples based on
survival predictor scores. 231 DLBCL samples were divided into quartiles based
on
their survival predictor scores. The survival predictor scores were calculated
by:
[0.586*(ABC DLBCL high gene expression signature value)] ¨ [0.468*(lymph node
gene expression signature value)] ¨ [(0.336*MHC Class II gene expression
signature
value)].
14
CA 2897828 2019-12-20

[0032] Figure 11: Kaplan-Meier plot of survival in DLBCL samples based on
survival predictor scores. 200 DLBCL samples were divided into quartiles based
on
their survival predictor scores. The survival predictor scores were calculated
by: [-
0.4337*(lymph node gene expression signature value)] + [0.09*(proliferation
gene
expression signature value)] ¨ [0.4144*(germinal center B-cell gene expression
signature value)] ¨ [0.2006*(MHC class II gene expression signature value)].
[0033] Figure 12: Kaplan-Meier plot of survival in DLBCL samples based on
survival predictor scores. 200 DLBCL samples were divided into quartiles based
on
their survival predictor scores. The survival predictor scores were calculated
by: I-
0.32*(lymph node gene expression signature value)] ¨ [0.176*(germinal center B-
cell
gene expression signature value)] ¨ [0.206*(MHC class II gene expression
signature
value)].
[0034] Figure 13: Kaplan-Meier plot of survival in MCL samples based on
survival
predictor scores. 21 MCL samples were divided into two equivalent groups based
on their survival predictor scores. The survival predictor scores were
calculated by:
1.66*(proliferation gene expression signature value).
[0035] Figure 14: Kaplan-Meier plot of survival in MCL samples based on
survival
predictor scores. 21 MCL samples were divided into two equivalent groups based
on their survival predictor scores. The survival predictor scores were
calculated by:
1.66*(proliferation gene expression signature value).
[0036] Figure 15: Predicting lymphoma type using Bayesian analysis. Bayes rule
can be used to determine the probability that an unknown sample belongs to a
first
lymphoma type rather than a second lymphoma type. A linear predictor score is
generated for the sample, and the probability that the sample belongs to the
first
CA 2897828 2019-12-20

lymphoma type is determined based on the distribution of linear predictor
scores
within the first and second lymphoma type.
[0037] Figure 16: Performance of MCL predictor model. Results of the gene-
expression based predictor model for MCL are shown for three models (MCL vs.
ABC, MCL vs. GCB, MCL vs. SLL). Performance is shown for both the training set
and the validation set.
[0038] Figure 17: Gene expression-based identification of DLBCL. Expression
levels for 27 genes in a subgroup predictor are shown for 274 DLBCL samples.
Expression levels are depicted according to the scale shown at the left. The
14
genes used to predict the DLBCL subgroups in the Affymetrix data set are
indicated
with asterisks. The probabilities that the DLBCL samples belong to the ABC or
GCB
subtypes are graphed at the top, and the DLBCL cases are arranged accordingly.
Cases belonging to either ABC or GCB with 90% or greater probability are
indicated.
[0039] Figure 18: Performance of DLBCL subtype predictor model. Assignments of
DLBCL samples to the ABC or GCB subtypes based on hierarchical clustering vs.
the predictor model disclosed herein are compared within the training,
validation,
and total set of samples.
[0040] Figure 19: Relationship of gene expression in normal B cell
subpopulations
to DLBCL subtypes. Relative gene expression in the indicated purified B cell
populations is depicted according to the scale in Figure 17. The P value of
the
difference in expression of these genes between the GCB and ABC DLBCL
subtypes is shown, and the subtype with the higher expression is shown. A.
DLBCL
subtype distinction genes that are more highly expressed in germinal center B
cells
than at other B cell differentiation stages. B. DLBCL subtype distinction
genes that
are more highly expressed in plasma cells than at other B cell differentiation
stages.
16
CA 2897828 2019-12-20

[0041] Figure 20: Identification of a PMBL gene expression signature. A.
Hierarchical clustering identified a set of 23 PMBL signature genes that were
more
highly expressed in most lymphomas with a clinical diagnosis of PMBL than in
lymphomas assigned to the GCB or ABC subtypes. Each row presents gene
expression measurements from a single Lymphochip microarray feature
representing the genes indicated. Each column represents a single lymphoma
biopsy sample. Relative gene expression is depicted according to the scale
shown.
B. Hierarchical clustering of the lymphoma biopsy samples based on expression
of
the PMBL signature genes identified in (A). A "core" cluster of lymphoma cases
was
identified that highly expressed the PMBL signature genes.
[0042] Figure 21: Development of a gene expression-based molecular diagnosis
of
PMBL. A. A PMBL predictor was created based on expression of the 46 genes
shown. Relative gene expression for each lymphoma biopsy sample is presented
according to the scale shown in Figure 20. The probability that each sample is
PMBL or DLBCL based on gene expression is shown at the top. B. The PMBL
predictor was used to classify 274 lymphoma samples as PMBL or DLBCL.
Prediction results are summarized on the right, and the relative gene
expression for
each case that was classified by the predictor as PMBL is shown on the left.
Average expression of each gene in samples classified as DLBCL is also shown.
The 20 genes listed are those represented on the Lymphochip that were more
highly
expressed in PMBL than in DLBCL. Not shown are eight genes from the PMBL
predictor that were more highly expressed in DLBCL than in PMBL.
[0043] Figure 22: Clinical characteristics of PMBL patients. Kaplan-Meier plot
of
overall survival in PMBL, GCB, and ABC patients after chemotherapy.
17
CA 2897828 2019-12-20

[0044] Figure 23: Optimization of gene number in lymphoma predictor. The
optimal
number of genes for inclusion in the lymphoma type predictor model is that
number
which generates a maximum t-statistic when comparing the LPS of two samples
from different lymphoma types.
[0045] Figure 24: LPS distribution among FL and DLBCL/BL samples. Standard
and proliferation LPSs for FL (x) and DLBCUBL (+) samples. Dotted lines
indicate
standard deviations from the fitted multivariate normal distributions.
[0046] Figure 25: Determination of cut-off points for lymphoma classification.
The
cut-off points between samples classified as DLBCL/BL, FL, or unclassified
were
optimized to minimize the number of samples classified as the wrong lymphoma
type. The optimal lower cut-off point was at q=0.49, while the optimal upper
cut-off
point was at q=0.84.
[0047] Figure 26: Division of LPSs among FL and DLBCL/FL samples. Illustration
of how the cut-off points described in Figure 25 divided the space between the
LPSs
of FL (x) and DLBCL/BL (+) samples.
[0048] Figure 27: Lymphoma classification results. Results of lymphoma
classification based on gene expression. 100% of SLL, MCL, and FH samples were
classified correctly, and only 3% of DLBCL/BL and FL samples were classified
incorrectly.
[0049] Figure 28: DLBCL subtype classification based on gene expression. None
of the ABC samples were classified as the wrong subtype, while only one of the
BL
samples was classified incorrectly. Of the GCB and PMBL samples, only 5% and
6%, respectively, were classified incorrectly.
[0050] Figure 29: Summary of chromosomal imbalances in DLBCL samples.
Chromosomal alterations in 224 untreated DLBCL samples were classified by gene
18
CA 2897828 2019-12-20

expression profiling. Each bar represents a chromosomal region gained or lost
in a
single sample. Bars on left side of ideogram represent losses of chromosomal
material. Bars on the right side of the ideogram represent gains of
chromosomal
material. Thick=bars on the right represent chromosomal gains exceeding the
cut-off
value of 1.5 in a large chromosomal region, and solid dots represent high-
level DNA
amplifications. A. Chromosomal alterations in GCB samples (n=87). B.
Chromosomal alterations in ABC samples (n=77). C. Chromosomal alterations in
PMBL samples (n=19). D. Diagram indicating frequencies of chromosomal
imbalances that distinguish between ABC, GCB, and PMBL. All differences were
statistically significant at p<0.05, with the exception of 12q12 gains
(P=0.059).
[0051] Figure 30: Influence of chromosomal gains and amplifications on locus-
specific gene expression levels. Changes in gene expression levels are
depicted for
each gene (averaged in each cohort) with regard to locus-specific genetic
status
(wild-type vs. gain vs. amplification). Genes are ordered according to their
chromosomal position. Gene locus information was obtained from the web site
for
Genes On Sequence Map (Homo sapiens built 33). For genes represented by more
than one element on the Lymphochip, the average expression of different clones
was calculated. The black bar on the left indicates the minimally gained
region in all
cases. Expression level comparisons were performed using the ANOVA test.
Genes with significant differences (P<0.01) are indicated.
[0052] Figure 31: Effect of chromosomal imbalances on gene expression
signatures. In each of the 4 panels, DLBCL cases are ordered according to
their
average expression of the following gene expression signatures: lymph node,
proliferation, T cell, and MHC class II. Samples with the chromosomal
abnormalities
shown on the right are marked. Correlations with a P-value < 0.05 are shown.
If
19
CA 2897828 2019-12-20

more than one cytoband in one chromosomal arm showed a P-value < 0.05, the
cytoband with the lowest p-value is displayed.
[0053] Figure 32: Impact of genomic gains of 3p11-p12 on survival of DLBCL
patients. Kaplan-Meier survival estimates of DLBCL patients with genomic gains
of
3p11-p12 in comparison to their stratification into survival quartiles based
on the
gene expression based outcome predictor model alone 3 (Q = Quartile)
(P=0.029).
[0054] Figure 32: Kaplan-Meier plot of survival in DLBCL samples based on
survival predictor scores. The survival predictor was calculated by:
[0.241*(proliferation gene expression signature value)] + [0.310*(BMP6)] ¨
[0.290*(germinal center B cell gene expression signature value)] ¨ [0.311*(MHC
class II gene expression signature value)] ¨ [0.249*(lymph node gene
expression
signature value)], with chromosome 3 gains involving the 3p11-p12 region
considered as an independent prognostic indicator.
[0055] Figure 33: Gene expression profiles of MCL signature genes in cyclin
D1-negative MCL cases. Expression profiles for six cyclin DI-negative MCL
cases were compared to those of ABC, GCB, PMBL, FL, MALT, SMZL, SLL,
and MCL. Median expression levels of the MCL signature genes are shown.
For the cyclin-D1 MCL cases, each column represents a single lymphoma
specimen and each row represents the level of expression of a single gene in
the MCL signature. Increased and decreased expression relative to the
median expression level according to the scale are shown over a four-fold
range. The lower panel shows expression levels of the D-type cyclins
according to the scale over a sixteen-fold range.
CA 2897828 2019-12-20

[0056] Figure 34: Cytologic features and expression of 0-type cyclins in
cyclin D1-
negative MCL. A. Typical MCL cytology (case 1) (hematoxylin and eosin stain,
original magnification X500). B. Cyclin D1 protein, showing only a rare non-
tumor
cell with nuclear staining (case 1). C. Cyclin D2 protein, showing strong
nuclear
staining of the tumor cells (case 2). D. Cyclin D3 protein, showing strong
nuclear
staining of the tumor cells (case 4) (immunoperoxidase stains, original
magnification
X400).
[0057] Figure 35: Molecular predictor of BL. A. Distinction between BL and
DLBCL using gene expression. Each row represents the expression level of a
gene and each column represents a lymphoma sample. Relative gene
expression levels are depicted according to the scale shown. Stage one
utilized c-myc and its target genes. Stage two utilized 100 additional genes
that distinguish BL from ABC, GCB, or PMBL. The panel includes only those
cases for which the pathology-based diagnosis and the gene expression-
based diagnosis agreed. B. Creation of an unbiased list of c-myc target genes
using RNA-interference experiments. The OCI-Ly10 DLBCL cell line was
transfected by electroporation with small interfering RNAs targeting the c-myc
gene, and gene expression was compared to that of control-transfected cells
by DNA microarray at the indicated times post-transfection. Downregulation of
c-myc and its targets is depicted according to the scale shown. C.
Performance of the gene expression-based predictor according to leave-one-
out cross-validation analysis. Samples submitted as BL or Burkitt-like
lymphoma were classified upon pathology review as classic and atypical BL.
Samples submitted as DLBCL were further classified by gene expression as
21
CA 2897828 2019-12-20

ABC, GCB, PMBL, or unclassified. D. Classification of samples submitted as
BL or Burkitt-like lymphoma that were reclassified as either DLBCL or high
grade lymphoma not otherwise specified. Also shown are samples submitted
and verified as high grade DLBCL. Those cases for which the pathology-
based diagnosis and the gene expression-based diagnosis disagreed (BL-
discrepant cases) are marked with an asterisk.
[0058] Figure 36: Performance of a molecular predictor of BL. A. Gene
expression
in the nine BL-discrepant cases for which the pathology-based diagnosis and
the
gene expression-based diagnosis were not in agreement. Expression of the BL-
predictor genes in these samples is compared to the average expression of
these
genes in BL and DLBCL. For each sample, immunophenotype, BCL2 mRNA and
protein expression, and t(14;18) FISH results are depicted at the bottom of
the
panel. Also shown is the probability that each sample is BL based on gene
expression. B. Expression of the BL-predictor genes in the six DLBCL samples
known to harbor a translocation involving the c-myc gene. Expression of these
genes in the samples is compared to the average expression in BL and DLBCL.
The probability that each sample is BL based on gene expression is shown.
[0059] Figure 37: Relative expression of gene expression signatures among
lymphoma subtypes. A. Average relative expression of c-myc and its target
genes for BL and each molecular subtype of DLBCL (ABC, GCB, and PMBL).
All expression data are shown over a 4-fold range. The scale is identical to
that in Figure 36. B. Expression of genes related to normal GC B cell
differentiation among the lymphoma subtypes. "BL-high" genes are expressed
at 2-fold or higher levels in BL compared to GCB (P<0.001). "BL-low" genes
22
CA 2897828 2019-12-20

are expressed at 2-fold or higher levels in GCB compared to BL (P<0.001).
The "BL¨GCB" genes are not differentially expressed between BL and GCB.
C. Relative expression among lymphoma types of genes that encode MHC
class-I proteins. D. Relative expression among the lymphoma types of genes
that are targets of the NF-KB signaling pathway. E. Average expression of
each of the four gene expression signatures among the lymphoma samples
according to their classification by the BL-predictor.
[0060] Figure 38: Survival analysis in BL and DLBCL. A. Kaplan-Meier plot of
overall survival for all patients with a gene expression-based diagnosis of
BL,
subdivided by treatment received. B. Kaplan-Meier plot of overall survival for
adults
with a gene expression-based diagnosis of BL, subdivided by treatment
received. C.
Kaplan-Meier plot of overall survival for all BL-discrepant patients,
subdivided by
treatment received. D. Kaplan-Meier plot of overall survival for all patients
assigned
to the three molecular subgroups of DLBCL.
DETAILED DESCRIPTION
[0061] The following description is merely intended to illustrate various
embodiments of the invention. As such, the specific modifications discussed
are not
to be construed as limitations on the scope of the invention. It will be
apparent to
one skilled in the art that various equivalents, changes, and modifications
may be
made without departing from the scope of the invention, and it is understood
that
such embodiments are to be included herein.
Abbreviations
[0062] The following abbreviations are used herein: ABC, activated B-cell-like
diffuse large B cell lymphoma; ASCT, autologous stem cell transplant; AWD,
alive
with disease; BL, Burkitt lymphoma; BM, bone marrow; CGH, comparative genome
23
CA 2897828 2019-12-20

hybridization; CHOP, cyclophosphamide, doxorubicine, vincristine, and
prednisone;
CI, confidence interval; CNS, central nervous system; COP, cyclophosphamide,
vincristine, and prednisone; CT, cycle threshold; DLBCL, diffuse large B-cell
lymphoma; DOD, dead of disease; ECOG, Eastern Cooperative Oncology Group;
EST, expressed sequence tag; FACS, fluorescence-activated cell sorting; FH,
follicular hyperplasia; FISH, fluorescence in situ hybridization; FL,
follicular
lymphoma; GC, germinal center; GCB, germinal center B-cell-like diffuse large
B cell
lymphoma; GI, gastrointestinal; IPI, International Prognostic Index; LPC,
lymphoplasmacytic lymphoma; LPS, linear predictor score; MALT, mucosa-
associated lymphoid tissue lymphomas; MCL, mantle cell lymphoma; MHC, major
histocompatibility complex; NA, not available or not applicable; NK, natural
killer;
NMZ, nodal marginal zone lymphoma; PB, peripheral blood; PCR, polymerase chain
reaction; PMBL, primary mediastinal B-cell lymphoma; PR, partial response;
PTLD,
post-transplant lymphoproliferative disorder; REAL, Revised European-American
Lymphoma; RPA, RNase protection assay; RQ-PCR, real-time quantitative PCR;
RR, relative risk of death; RT-PCR, reverse transcriptase polymerase chain
reaction;
SAGE, serial analysis of gene expression; SLL, small lymphocytic lymphoma;
SMZL,
splenic marginal zone lymphoma; WHO, World Health Organization.
Definitions
[0063] The term "Iymphoproliferative disorder" as used herein refers to any
tumor
of lymphocytes, and may refer to both malignant and benign tumors. The terms
"lymphoma" and "lymphoid malignancy" as used herein refer specifically to
malignant tumors derived from lymphocytes and lymphoblasts. Examples of
lymphomas include, but are not limited to, follicular lymphoma (FL), Burkitt
lymphoma (BL), mantle cell lymphoma (MCL), follicular hyperplasia (FH), small
cell
24
CA 2897828 2019-12-20

lymphocytic lymphoma (SLL), mucosa-associated lymphoid tissue lymphoma
(MALT), splenic lymphoma, multiple myeloma, lymphoplasmacytic lymphoma, post-
transplant lymphoproliferative disorder (PTLD), lymphoblastic lymphoma, nodal
marginal zone lymphoma (NMZ), germinal center B cell-like diffuse large B cell
lymphoma (GCB), activated B cell-like diffuse large B cell lymphoma (ABC) and
primary mediastinal B cell lymphoma (PMBL).
[0064] The phrase "lymphoma type" (or simply "type") as used herein refers to
a
diagnostic classification of a lymphoma. The phrase may refer to a broad
lymphoma
class (e.g., DLBCL, FL, MCL, etc.) or to a subtype or subgroup falling within
a broad
lymphoma class (e.g., GCB DLBCL, ABC DLBCL).
[0065] The phrase "gene expression data" as used herein refers to information
regarding the relative or absolute level of expression of a gene or set of
genes in a
cell or group of cells. The level of expression of a gene may be determined
based on
the level of RNA, such as mRNA, encoded by the gene. Alternatively, the level
of
expression may be determined based on the level of a polypeptide or fragment
thereof encoded by the gene. "Gene expression data" may be acquired for an
individual cell, or for a group of cells such as a tumor or biopsy sample.
[0066] The term "microarray," "array," or "chip" refers to a plurality of
nucleic acid
probes coupled to the surface of a substrate in different known locations. The
substrate is preferably solid. Microarrays have been generally described in
the art
in, for example, U.S. Patent Nos. 5,143,854 (Pirrung), 5,424,186 (Fodor),
5,445,934
(Fodor), 5,677,195 (Winkler), 5,744,305 (Fodor), 5,800,992 (Fodor), 6,040,193
(Winkler), and Fodor et al. 1991. Light-directed, spatially addressable
parallel
chemical synthesis. Science, 251:767-777.
CA 2897828 2019-12-20

[0067] The term "gene expression signature" or "signature" as used herein
refers to
a group of coordinately expressed genes. The genes making up this signature
may
be expressed in a specific cell lineage, stage of differentiation, or during a
particular
biological response. The genes can reflect biological aspects of the tumors in
which
they are expressed, such as the cell of origin of the cancer, the nature of
the non-
malignant cells in the biopsy, and the oncogenic mechanisms responsible for
the
cancer (Shaffer 2001). Examples of gene expression signatures include lymph
node
(Shaffer 2001), proliferation (Rosenwald 2002), MHC class II, ABC DLBCL high,
B-
cell differentiation, T-cell, macrophage, immune response-1, immune response-
2,
and germinal center B cell.
[0068] The phrase "survival predictor score" as used herein refers to a score
generated by a multivariate model used to predict survival based on gene
expression. A subject with a higher survival predictor score is predicted to
have
poorer survival than a subject with a lower survival predictor score.
[0069] The term "survival" as used herein may refer to the probability or
likelihood
of a subject surviving for a particular period of time. Alternatively, it may
refer to the
likely term of survival for a subject, such as expected mean or median
survival time
for a subject with a particular gene expression pattern.
[0070] The phrase "linear predictor score" or "LPS" as used herein refers to a
score
that denotes the probability that a sample belongs to a particular lymphoma
type.
An LPS may be calculated using an equation such as:
LPS(S) =
,EG
where Si is the expression of gene] from gene set G in a sample S, and ti is a
scale
factor representing the difference in expression of gene] between a first
lymphoma
26
CA 2897828 2019-12-20

type and a second lymphoma type. Alternatively, a linear predictor score may
be
generated by other methods including but not limited to linear discriminant
analysis
(Dudoit 2002), support vector machines (Furey 2000), or shrunken centroids
(Tibshirani 2002)
[0071] The phrase "scale factor" as used herein refers to a factor that
defines the
relative difference in expression of a particular gene between two samples. An
example of a scale factor is a t-score generated by a Student's t-test.
[0072] The phrase "lymphoma subject," wherein "lymphoma" is a specific
lymphoma type (e.g., "follicular lymphoma subject"), may refer to a subject
that has
been diagnosed with a particular lymphoma by any method known in the art or
discussed herein. This phrase may also refer to a subject with a known or
suspected predisposition or risk of developing a particular lymphoma type.
[0073] The gene expression profile of a cancer cell or biopsy sample at a
specific
timepoint may provide the basis for better classification of cancer subtypes,
more
accurate prediction of cancer survival, and more specifically tailored
therapies.
Disclosed herein are a variety of methods for identifying, diagnosing, and/or
classifying a lymphoma, lymphoid malignancy, or lymphoproliferative disorder
based
on its gene expression patterns. Also disclosed are methods for predicting
survival
in a subject diagnosed with a particular lymphoma type or subtype using gene
expression data. The information obtained using these methods will be useful
in
evaluating the optimal therapeutic approach to be employed for a particular
subject
suffering from cancer.
[0074] The pattern of expression of a particular gene is closely connected to
the
biological role and effect of its gene product. For this reason, the
systematic study
of variations in gene expression provides an alternative approach for linking
specific
27
CA 2897828 2019-12-20

genes with specific diseases and for recognizing heritable gene variations
that are
important for immune function. For example, allelic differences in the
regulatory
region of a gene may influence the expression levels of that gene. An
appreciation
for such quantitative traits in the immune system may help elucidate the
genetics of
autoimmune diseases and lymphoproliferative disorders.
[0075] Genes that encode components of the same multi-subunit protein complex
are often coordinately regulated. Coordinate regulation is also observed among
genes whose products function in a common differentiation program or in the
same
physiological response pathway. Recent application of gene expression
profiling to
the immune system has shown that lymphocyte differentiation and activation are
accompanied by parallel changes in expression among hundreds of genes. Gene
expression databases may be used to interpret the pathological changes in gene
expression that accompany autoimmunity, immune deficiencies, cancers of immune
cells and of normal immune responses.
[0076] Scanning and interpreting large bodies of relative gene expression data
is a
formidable task. This task is greatly facilitated by algorithms designed to
organize
the data in a way that highlights systematic features, and by visualization
tools that
represent the differential expression of each gene as varying intensities and
hues of
color (Eisen 1998). The development of microarrays, which are capable of
generating massive amounts of expression data in a single experiment, has
greatly
increased the need for faster and more efficient methods of analyzing large-
scale
expression data sets. In order to effectively utilize microarray gene
expression data
for the identification and diagnosis of lymphoma and for the prediction of
survival in
lymphoma patients, new algorithms must be developed to identify important
information and convert it to a more manageable format. In addition, the
28
CA 2897828 2019-12-20

microarrays used to generate this data should be streamlined to incorporate
probe
sets that are useful for diagnosis and survival prediction. Disclosed herein
are
various methods and compositions that address both of these issues.
[0077] Mathematical analysis of gene expression data is a rapidly evolving
science
based on a rich mathematics of pattern recognition developed in other contexts
(Kohonen 1997). Mathematical analysis of gene expression generally has three
goals. First, it may be used to identify groups of genes that are coordinately
regulated within a biological system. Second, it may be used to recognize and
interpret similarities between biological samples on the basis of similarities
in gene
expression patterns. Third, it may be used to recognize and identify those
features
of a gene expression pattern that are related to distinct biological processes
or
phenotypes.
[0078] Mathematical analysis of gene expression data often begins by
establishing
the expression pattern for each gene on an array across n experimental
samples.
The expression pattern of each gene can be represented by a point in n-
dimensional
space, with each coordinate specified by an expression measurement in one of
the
n samples (Eisen 1998). A clustering algorithm that uses distance metrics can
then
be applied to locate clusters of genes in this n-dimensional space. These
clusters
indicate genes with similar patterns of variation in expression over a series
of
experiments. Clustering methods that have been applied to microarray data in
the
past include hierarchical clustering (Eisen 1998), self-organizing maps (SOMs)
(Tamayo 1999), k-means (Tavazoie 1999), and deterministic annealing (Alon
1999).
A variety of different algorithms, each emphasizing distinct orderly features
of the data, may be required to glean the maximal biological insight from a
set of
samples (Alizadeh 1998). One such algorithm, hierarchical clustering, begins
by
29
CA 2897828 2019-12-20

determining the gene expression correlation coefficients for each pair of the
n genes
studied. Genes with similar gene expression correlation coefficients are
grouped
next to one another in a hierarchical fashion. Generally, genes with similar
expression patterns under a particular set of conditions encode protein
products that
play related roles in the physiological adaptation to those conditions. Novel
genes of
unknown function that are clustered with a large group of functionally related
genes
are likely to participate in the same biological process. Likewise, the other
clustering
methods mentioned herein may also group genes together that encode proteins
with
related biological function.
[0079] Gene expression maps may be constructed by organizing gene expression
data from multiple samples using any of the various clustering algorithms
outlined
herein. The ordered tables of data may then be displayed graphically in a way
that
allows researchers and clinicians to assimilate both the choreography of gene
expression on a broad scale and the fine distinctions in expression of
individual
genes.
[0080] In such a gene expression map, genes that are clustered together
reflect a
particular biological function, and are termed gene expression signatures
(Shaffer
2001). One general type of gene expression signature includes genes that are
characteristically expressed in a particular cell type or at a particular
stage of cellular
differentiation or activation. Another general type of gene expression
signature
includes genes that are regulated in their expression by a particular
biological
process such as proliferation, or by the activity of a particular
transcription factor or
signaling pathway.
[0081] The pattern of gene expression in a biological sample provides a
distinctive
and accessible molecular picture of its functional state and identity (DeRisi
1997;
CA 2897828 2019-12-20

Cho 1998; Chu 1998; Holstege 1998; Spellman 1998). Each cell transduces
variations in its environment, internal state, and developmental state into
readily
measured and recognizable variations in its gene expression patterns. Two
different
samples that have related gene expression patterns are therefore likely to be
biologically and functionally similar to one another. Some biological
processes are
reflected by the expression of genes in a specific gene expression signature,
as
described above. The expression of a specific gene expression signature in a
sample can provide important biological insights into its cellular composition
and the
function of various intracellular pathways within those cells.
[0082] The present invention discloses a variety of gene expression signatures
related to the clinical outcome of lymphoma patients. While several of these
signatures share a name with a previously disclosed signature, each of the
gene
expression signatures disclosed herein comprises a novel combination of genes.
For example, the lymph node signature disclosed herein includes genes encoding
extracellular matrix components and genes that are characteristically
expressed in
macrophage, NK, and T cells (e.g., a-Actinin, collagen type Ill a 1,
connective tissue
growth factor, fibronectin, KIAA0233, urokinase plasminogen activator). The
proliferation signature includes genes that are characteristically expressed
by cells
that are rapidly multiplying or proliferating (e.g., c-myc, E21G3, NPM3,
BMP6). The
MHC class II signature includes genes that interact with lymphocytes in order
to
allow the recognition of foreign antigens (e.g., HLA-DPa, HLA-DQa, HLA-DRa,
HLA-
DR6). The immune response-1 signature includes genes encoding T cell markers
(e.g., CD7, CD8B1, ITK, LEF1, STAT4), as well as genes that are highly
expressed
in macrophages (e.g., ACTN1, TNFSF13B). The immune response-2 signature
includes genes known to be preferentially expressed in macrophages and/or
31
CA 2897828 2019-12-20

dendritic cells (e.g., TLR5, FCGR1A, SEPT10, LGMN, C3AR1). The germinal
center B cell signature includes genes known to be overexpressed at this stage
of B
cell differentiation (e.g., MME, MEF2C, BCL6, LM02, PRSPAP2, MBD4, EBF,
MYBL1).
[0083] Databases of gene expression signatures have proven quite useful in
elucidating the complex gene expression patterns of various cancers. For
example,
expression of genes from the germinal center B-cell signature in a lymphoma
biopsy
suggests that the lymphoma is derived from this stage of B cell
differentiation. In the
same lymphoma biopsy, the expression of genes from the T cell signature can be
used to estimate the degree of infiltration of the tumor by host T cells,
while the
expression of genes from the proliferation signature can be used to quantitate
the
tumor cell proliferation rate. In this manner, gene expression signatures
provide an
"executive summary" of the biological properties of a tumor specimen. Gene
expression signatures can also be helpful in interpreting the results of a
supervised
analysis of gene expression data. Supervised analysis generates a long list of
genes with expression patterns that are correlated with survival. Gene
expression
signatures can be useful in assigning these "predictive" genes to functional
categories. In building a multivariate model of survival based on gene
expression
data, this functional categorization helps to limit the inclusion of multiple
genes in the
model that measure the same aspect of tumor biology.
[0084] Gene expression profiles can be used to create multivariate models for
predicting survival. The methods for creating these models are called
"supervised"
because they use clinical data to guide the selection of genes to be used in
the
prognostic classification. For example, a supervised method might identify
genes
with expression patterns that correlate with the length of overall survival
following
32
CA 2897828 2019-12-20

chemotherapy. The general method used to create a multivariate model for
predicting survival may utilize the following steps:
1. Identify genes with expression patterns that are univariately associated
with a particular clinical outcome using a Cox proportional hazards
model. Generally, a univariate p-value of <0.01 is considered the cut-off
for significance. These genes are termed "predictor" genes.
2. Within a set of predictor genes, identify gene expression signatures.
3. For each gene expression signature that is significantly associated with
survival, average the expression of the component genes within this
signature to generate a gene expression signature value.
4. Build a multivariate Cox model of clinical outcome using the gene
expression signature values.
5. If possible, include additional genes in the model that do not belong to a
gene expression signature but which add to the statistical power of the
model.
This approach has been utilized in the present invention to create novel
survival
prediction models for FL, DLBCL, and MCL. Each of these models generates a
survival predictor score, with a higher score being associated with worse
clinical
outcome. Each of these models may be used separately to predict survival.
Alternatively, these models may be used in conjunction with one or more other
models, disclosed herein or in other references, to predict survival.
[0085] A first FL survival predictor was generated using gene expression data
obtained using Affymetrix U133A and U133B microarrays. This predictor
incorporated immune response-1 and immune response-2 gene expression
33
CA 2897828 2019-12-20

signatures. Fitting the Cox proportional hazards model to the gene expression
signature values obtained from these signatures resulted in the following
model:
Survival predictor score = [(2.71*immune response-2 gene expression
signature value)] ¨ [(2.36 x immune response-1 gene expression
signature value)].
[0086] A second FL survival predictor was generated using gene expression data
obtained using Affymetrix U133A and U133B microarrays. This predictor
incorporated macrophage, T-cell, and B-cell differentiation gene expression
signatures. Fitting the Cox proportional hazards model to the gene expression
signature values obtained from these signatures resulted in the following
model:
Survival predictor score = [2.053*(macrophage gene expression
signature value)] ¨ [2.344*(T-cell gene expression signature value)] ¨
[0.729*(B-cell differentiation gene expression signature value)].
[0087] A third FL survival predictor was generated using gene expression data
obtained using the Lymph Dx microarray. This predictor incorporated
macrophage,
T-cell, and B-cell differentiation gene expression signatures. Fitting the Cox
proportional hazards model to the gene expression signature values obtained
from
these signatures resulted in the following model:
Survival predictor score = [1.51*(macrophage gene expression
signature
value)] ¨ [2.11*(T-cell gene expression signature value)] ¨ [0.505*(B-
cell differentiation gene expression signature value)].
[0088] A first DLBCL survival predictor was generated using gene expression
data
obtained using Affymetrix U133A and U133B microarrays. This predictor
incorporated ABC DLBCL high, lymph node, and MHC class II gene expression
34
CA 2897828 2019-12-20

signatures. Fitting the Cox proportional hazards model to the gene expression
signature values obtained from these signatures resulted in the following
model:
Survival predictor score = [0.586*(ABC DLBCL high gene expression
signature value)] ¨ [0.468*(lymph node gene expression signature
value)] ¨ 0.336*(MHC class ll gene expression signature value)].
[0089] A second DLBCL survival predictor was generated using gene expression
data obtained using the Lymph Dx microarray. This predictor incorporated lymph
node, proliferation, germinal center B-cell, and MHC class II gene expression
signatures. Fitting the Cox proportional hazards model to the gene expression
signature values obtained from these signatures resulted in the following
model:
Survival predictor score = [-0.4337*(lymph node gene expression
signature value)] + [0.09*(proliferation gene expression signature
value)] ¨ [0.4144*(germinal center B-cell gene expression signature
value)] ¨[0.2006*(MHC class II gene expression signature value)].
[0090] A third DLBCL survival predictor was generated using gene expression
data
obtained using the Lymph Dx microarray. This predictor incorporated lymph
node,
germinal center B cell, and MHC class II gene expression signatures. Fitting
the
Cox proportional hazards model to the gene expression signature values
obtained
from these signatures resulted in the following model:
Survival predictor score = [-0.32*(lymph node gene expression
signature value)] ¨ [0.176*(germinal center B-cell gene expression
signature value)] ¨ [0.206*(MHC class II gene expression signature
value)].
[0091] A method of refining a DLBCL survival predictor was developed based on
analysis of characteristic chromosomal alterations in DLBCL cases. Malignant
CA 2897828 2019-12-20

lymphomas are generally characterized by distinctive recurrent primary
chromosomal translocations such as the t(11;14) or t(14;18) in MCL and FL,
respectively. By identifying genomic imbalances, comparative genomic
hybridization
(CGH) has the potential to detect less well-characterized chromosomal
aberrations
in lymphomas that may play an important role in development and progression of
the
disease. In DLBCL, previous cytogenetic studies have identified a plethora of
clonal
chromosomal aberrations, some of which are associated with particular
morphological or clinical manifestations (Yunis 1989; Monni 1996; Rao 1998;
Cigudosa 1999; Schlegelberger 1999; Berglund 2002; Dave 2002; Nanjangud 2002;
Bea 2004).
[0092] CGH was used to identify chromosomal alterations in 224 DLBCL samples
that had previously been broken into subtypes based on gene expression data
obtained using a Lymphochip microarray. Chromosomal alterations were observed
in 164 of the 224 cases. Certain alterations were differentially distributed
among
subtypes. For example, gains of chromosome arm 3q were observed in around a
fourth of ABC samples, but were never observed in GCB samples and were
observed only once in PMBL samples. Similarly, gains of 18q21-q22 were
observed
in around a third of ABC samples, but were observed in only 10% and 16% of GCB
and PMBL samples, respectively. Genomic gains of 3q and 18q have previously
been correlated with shorter survival in DLBCL patients (Bea 2004). The
present
findings provide a clear explanation for this observation, namely that both
abnormalities are statistically associated with ABC, which has a worse
prognosis
than other DLBCL subtypes (Alizadeh 2000; Rosenwald 2002; Rosenwald 2003b).
Alternatively, these genetic alterations may themselves contribute at least in
part to
the ABC gene expression phenotype and its inferior prognosis.
36
CA 2897828 2019-12-20

[0093] Other chromosomal abnormalities were found to occur more frequently in
one DLBCL subtype than another, but without being restricted to a single
subtype.
For example, deletions of 6q21-q22 occurred in 40% was observed in 40% of ABC
samples and 22% of GCB samples, but never in PMBL samples. Gains and
amplifications of 12cen-q15 were observed most frequently in GCB samples, but
were also observed at a low frequency in both ABC and PMBL samples. These
results suggest that certain oncogenic pathways are shared by various DLBCL
subtypes, but nonetheless are more frequently utilized in different subtypes.
[0094] The distinct patterns of genomic alterations observed across different
DLBCL subtypes are consistent with recent studies showing a correlation
between
gene copy number changes and expression of genes located within the involved
genomic regions (Phillips 2001; Virtaneva 2001; Hyman 2002; Pollack 2002;
Orsetti
2004).
[0095] The relationship between chromosome gains/amplifications and the
expression profile of genes located within the gained/amplified regions was
examined by RQ-PCR for four chromosomal regions commonly overrepresented in
GCB and ABC tumors: 2p14-p16, 3q27-qter, 12q12-q15, and 18q21-q22. A strong
impact of genomic gains and amplifications on gene expression was observed. 25-
75% of genes located within these chromosomal segments were overexpressed in
those tumors with increased DNA copy number.
[0096] For many genes, expression levels were significantly higher in samples
with
gains or amplifications versus samples with normal DNA profiles, which
suggests a
direct effect of copy number on mRNA expression levels. However, not all genes
in
overrepresented chromosomal regions were more highly expressed. This suggests
that either the individual genes were not amplified or that the functional
background
37
CA 2897828 2019-12-20

of the cell was not appropriate for expression of the gene. For example, REL
was
significantly overexpressed in GCB samples with overrepresentation of 2p14-
p16,
but not in ABC samples with overrepresentation of this region. Quantitative
PCR
analysis confirmed that the REL locus was amplified in virtually all GCB
samples
with overrepresentation of the 2p14-p16 region. However, REL was not amplified
in
any of the ABC samples with gains or amplifications of 2p14-p16, indicating
that
genes other than REL may be targeted by 2p14-p16 gains in ABC. Interestingly,
mRNA expression of BCL11A, which is located very close to REL, was not
influenced by 2p14-p16 gains in GCB or ABC samples, despite the fact that
quantitative PCR analysis showed amplification and gains of the BCL11A locus
in
both subtypes.
[0097] Gene expression studies showed that chromosomal alterations in DLBCL
cases are correlated with expression of certain gene expression signatures.
Unexpectedly, this correlation was seen not only in gene expression signatures
that
reflect variation within malignant cells (proliferation and MHC class II
signatures), but
also in gene expression signatures that reflect the nature of non-malignant
DLBCL
tumors (T cell and lymph node signatures). The proliferation signature, which
is
more highly expressed in proliferating than in quiescent cells, was increased
in
DLBCL samples with genomic loss in 6q21 and gains in several bands of
chromosome 3. The MHC class II signature, which reflects the coordinate
regulation
of all MHC class II genes in malignant DLBCL cells, was decreased in DLBCL
samples with gains of 3p11-p12. The T cell signature, which reflects the
infiltration
of tumors by T cells, was decreased in DLBCL samples with gains of cytobands
in
chromosomes 7, 11, 12, and X or losses in 6q and 17p. The lymph node
signature,
which reflects a host response characterized by abundant expression of
extracellular
38
CA 2897828 2019-12-20

matrix components and infiltration of tumors with immune cells other than T
cells,
was increased in DLBCL 'samples with Xp21 gains and decreased in samples with
gains in several cytobands of chromosome 3.
[0098] Previous studies have suggested that specific genetic alterations are
relevant to predicting survival in DLBCL cases (Yunis 1989; Bea 2004).
However,
the present study shows that only gains in certain regions of chromosome 3 are
significantly associated with inferior survival after adjusting for multiple
variable
comparisons. Specifically, gains involving 3p11-p12 were found to have
prognostic
value that was statistically independent of previously defined DLBCL survival
predictors based on optimal gene expression-based models (Rosenwald 2002).
This information can be used to create a refined DLBCL survival predictor. For
example, a survival predictor score may be calculated using an equation such
as:
Survival predictor score = [0.241*(proliferation gene expression
signature value)] + [0.310*(BMP6 expression value)] ¨
[0.290*(germinal center B cell gene expression signature value)] ¨
[0.311*(MHC class II gene expression signature value)] ¨
[0.249*(lymph node gene expression signature value)],
wherein a higher survival predictor score is associated with worse survival
prognosis.
The DLBCL sample may then be assayed for gains or amplifications in the 3p11-
p12
using any method, such as for example CGH. The identification of such gains or
amplifications corresponds to a decrease in survival, and may be used to
adjust the
survival predictor score accordingly, i.e., to increase the survival predictor
score).
Alternatively, Identification of a gain or amplification of 3p11-p12 may be
used as a
stand-alone indicator of worse survival prognosis for a DLBCL patient in the
absence
of additional gene expression data.
39
CA 2897828 2019-12-20

[0099] An MCL survival predictor was generated using gene expression data
obtained using Affymetrix U133A, Affymetrix U133B, and Lymph Dx microarrays.
This predictor incorporated a proliferation gene expression signature. Fitting
the
Cox proportional hazards model to the gene expression signature values
obtained
from these signatures resulted in the following model:
Survival predictor score = [1.66*(proliferation gene expression
signature value)].
[00100] Gene expression data can also be used to diagnose and identify
lymphoma
types. In an embodiment of the present invention, a statistical method based
on
Bayesian analysis was developed to classify lymphoma specimens according to
their
gene expression profiles. This method does not merely assign a tumor to a
particular lymphoma type, but also determines the probability that the tumor
belongs
to that lymphoma type. Many different methods have been formulated to predict
cancer subgroups (Golub 1999; Ramaswamy 2001; Dudoit 2002; Radmacher 2002).
These methods assign tumors to one of two subgroups based on expression of a
set
of differentially expressed genes. However, they do not provide a probability
of
membership in a subgroup. By contrast, the method disclosed herein used Bayes'
rule to estimate this probability, thus allowing one to vary the probability
cut-off for
assignment of a tumor to a particular subgroup. In tumor types in which
unknown
additional subgroups may exist, the present method allows samples that do not
meet
the gene expression criteria of known subgroups to fall into an unclassified
group
with intermediate probability. A cancer subgroup predictor of the type
described
herein may be used clinically to provide quantitative diagnostic information
for an
individual cancer patient. This information can in turn be used to provide a
predictor
of treatment outcome for a particular cancer patient.
CA 2897828 2019-12-20

[00101] For any two lymphoma types A and B, there is a set of genes with
significantly higher expression in type A than type B, and a set of genes with
significantly lower expression in type A than in type B. By observing the
expression
of these genes in an unknown sample, it is possible to determine to which of
the two
types the sample belongs. Evaluating the likelihood that a particular sample
belongs
to one or the other lymphoma type by Bayesian analysis may be done using the
following steps:
1. Identify those genes that are most differentially expressed between the two
lymphoma types. This can be done by selecting those genes with the
largest t-statistic between the two lymphoma types. The genes in this step
may be subdivided into gene expression signatures in certain cases, with
genes from each signature analyzed separately.
2. Create a series of linear predictor score (LPS) for samples belonging to
either lymphoma type.
3. Evaluate the LPS for each sample in a training set, and estimate the
distribution of these scores within each lymphoma type according to a
normal distribution.
4. Use Bayes' rule to evaluate the probability that each subsequent sample
belongs to one or the other lymphoma type.
If only two types of lymphoma are being distinguished, then a single
probability
score is sufficient to discriminate between the two types. However, if more
than two
lymphoma types are being distinguished, multiple scores will be needed to
highlight
specific differences between the types.
(00102]A novel microarray termed the Lymph Dx microarray is disclosed herein
for
the identification and diagnosis of various lymphoma types. The Lymph Dx
41
CA 2897828 2019-12-20

microarray contains cDNA probes corresponding to approximately 2,653 genes,
fewer than the number seen on microarrays that have been used previously for
lymphoma diagnosis. The reduced number of probes on the Lymph Dx microarray
is the result of eliminating genes that are less useful for the identification
of
lymphoma types and predicting clinical outcome. This reduction allows for
simplified
analysis of gene expression data. The genes represented on the Lymph Dx
microarray can be divided into four broad categories: 1,101 lymphoma predictor
genes identified previously using the Affymetrix U133 microarray, 171 outcome
predictor genes, 167 new genes not found on the Affymetrix U133 microarray,
and
1,121 named genes. A list of the probe sets on the Lymph Dx microarray is
presented in Table 2, contained in the file
"Table_0002_LymphDx_Probe_List.txt."
[00103] Gene expression data obtained using the Lymph Dx microarray may be
used to identify and classify lymphomas using Bayesian analysis using a
strategy
similar to that set forth above. In certain embodiments, this strategy may
include
additional steps designed to optimize the number of genes used and the cut-off
points between lymphoma types. A general overview of such a method is
presented
in Figure 1. In one example of the method, each gene represented on the Lymph
Dx microarray was placed into one of three gene-list categories based on its
correlation with the lymph node or proliferation gene expression signatures:
lymph
node, proliferation, or standard. These signatures were identified by
clustering of
the DLBCL cases using hierarchical clustering and centroid-correlation of
0.35.
Standard genes were those with expression patterns that did not correlate
highly
with expression of the lymph node or proliferation signatures. Lymph Dx gene
expression data was first used to identify samples as FL, MCL, SLL, FH, or
DLBCL/BL, then to identify DLBCL/BL samples as ABC, GCB, PMBL, or BL. For
42
CA 2897828 2019-12-20

each stage, a series of pair-wise models was created, with each model
containing a
different pair of lymphoma types (e.g., FL vs. MCL, SLL vs. FH, etc.). For
each pair,
the difference in expression of each gene on the microarray was measured, and
a t-
statistic was generated representing this difference. Genes from each gene-
list
category were ordered based on their t-statistic, and those with the largest t-
statistics
were used to generate a series of LPSs for samples belonging to either
lymphoma
type. The number of genes used to generate the LPSs was optimized by repeating
the calculation using between five and 100 genes from each gene-list category.
The
number of genes from each category used in the final LPS calculation was that
which gave rise to the largest difference in LPS between the two lymphoma
types.
Once the number of genes in each gene-list category was optimized, four
different
LPSs were calculated for each sample. The first included genes from the
standard
gene-list category only, the second included genes from the proliferation and
standard gene-list categories, the third included genes from the lymph node
and
standard gene-list categories, and the fourth included genes from all three
categories. The probability q that a sample X belongs to the first lymphoma
type of
a pair-wise model can then be calculated using an equation:
q = o(Lps(x);
OLPS(X); /2õa,)+0(LPS(X); /2' 02)
LPS(X) is the LPS for sample X, cb(x; p, a) is the normal density function
with
mean and standard deviation a, A, and .5-, are the mean and variance of the
LPSs
for samples belonging to the first lymphoma type, and anda, are the mean
and
variance of the LPSs for samples belonging to the second lymphoma type.
Samples
with high q values were classified as the first lymphoma type, samples with
low q
values were classified as the second lymphoma type, and samples with middle
43
CA 2897828 2019-12-20

range q values were deemed unclassified. To determine the proper cut-off point
between high, low, and middle q values, every possible cut-off point between
adjacent samples was analyzed by an equation:
3.99 * [(% of type 1 misidentified as type 2) + (% of type 2 misidentified
as type 1)] + [(% of type 1 unclassified) + (% of type 2 misidentified)].
This equation was used to favor the assignment of a sample to an
"unclassified"
category rather than to an incorrect lymphoma type. The final cut-off points
were
those which minimized this equation. The coefficient of 3.99 was chosen
arbitrarily
to allow an additional classification error only if the adjustment resulted in
four or
more unclassified samples becoming correctly classified. The coefficient can
be
varied to achieve a different set of trade-offs between the number of
unclassified
and misidentified samples.
[00104] To ensure that the accuracy of the model was not a result of
overfilling,
each model was validated by leave-one-out cross-validation. This entailed
removing
each sample of known lymphoma type from the data one at a time, and then
determining whether the model could predict the missing sample. This process
confirmed the accuracy of the prediction method.
[00105] Bayesian analysis has been used herein to classify lymphoma samples as
DLBCL or BL based on gene expression data. BL is characterized by a high
degree
of proliferation and deregulation of the c-myc gene (Jaffe 2001). The
diagnostic
distinction between BL and DLBCL is critically important because there are
significant differences in their clinical management. Lower-dose chemotherapy
regimens typically used to treat DLBCL (e.g., CHOP) are not adequate to treat
BL
(Bishop 2000; Butler 1993), which requires intensive chemotherapy regimens
(Pees
1992; Magrath 1996; Thomas 1999; Mead 2002; Divine 2005). Furthermore,
44
CA 2897828 2019-12-20

prophylactic intrathecal chemotherapy or systemic chemotherapy that crosses
the
blood-brain barrier, which are unnecessary in most cases of DLBCL, are
essential in
the treatment of BL due the high risk of central nervous system involvement
(Soussain 1995; Bishop 2000).
[00106] The diagnosis of BL relies on morphology, immunophenotype, and
cytogenetics (Jaffe 2001). However, DLBCL and BL can have overlapping
morphology and immunophenotype, and the characteristic t(8;14) translocation
of
BL (Neri 1988; Gerbitz 1999; Hecht 2000) is also found in 5-10% of DLBCL cases
(Kramer 1998). Since DLBCL is over 20 times more common than BL (Morton
2005), most aggressive lymphomas with t(8; 1 4) are not BL. Thus, the
distinction
between BL and DLBCL can be difficult and may lead to incorrect clinical
decisions
and adverse outcomes.
[00107] The term Burkitt-like lymphoma has been used to refer to cases that
have
some features in common with BL. However, the most recent WHO guidelines
(Jaffe 2005) have eliminated Burkitt-like lymphoma as a separate diagnostic
category. Burkitt-like lymphoma is now synonymous with the term atypical BL,
which
is reserved for those cases that share the genetic abnormality and
immunophenotype of BL but have atypical morphology. It is not clear whether
atypical BL is biologically difference from BL or if it merely represents a
morphologic
variant.
[00108]A gene expression-based predictor of BL that diagnoses classic BL with
100% accuracy and distinguishes it from DLBCL has been developed.
Surprisingly,
eight cases that were given a pathological diagnosis of DLBCL were similar to
BL by
gene expression and had other molecular and clinical characteristics of BL.
CA 2897828 2019-12-20

[00109] Compared to DLBCL, BL was found to have high expression of the c-myc
target gene expression signature and the GC B cell gene expression signature,
and
low expression of the NF-KB target gene expression signature and the MHC class
I
gene expression signature. A number of aggressive lymphomas that had been
classified as DLBCL by an expert panel of hematopathologists (n=8) were
reclassified as BL based on gene expression analysis. All eight of these cases
had
c-myc translocations and resembled BL with respect to all four gene expression
signatures that distinguished BL from DLBCL, suggesting that these cases
represent
BL that cannot be reliably diagnosed by current methods. Consistent with
previous
studies (Butler 1993; Magrath 1996; Smeland 2004), patients classified as BL
by
gene expression had poor outcome with lower dose chemotherapy regimens, yet
could be cured with intensive regimes.
[00110] The translocation of the c-myc gene and its consequent deregulation is
a
key oncogenic event in the genesis of BL and, accordingly, expression of the c-
myc
target gene expression signature distinguished BL from DLBCL. However, c-myc
translocations also occur in 5-10% of DLBCL. It is therefore noteworthy that
the
gene expression-based predictor disclosed herein did not classify any of six
DLBCL
cases bearing a c-myc translocation as BL. Thus, c-myc deregulation by
translocation and the attendant overexpression of c-myc target genes are not
sufficient to create the phenotype of BL. Rather, it is likely that additional
differences
in the molecular pathogenesis of BL and DLBCL contribute to their clinical
differences.
[00111] In keeping with this notion, BL and DLBCL were found to differ in the
expression of three gene expression signatures in addition to the c-myc target
gene
signature. Both BL and GCB are thought to originate from a germinal center B
cell
46
CA 2897828 2019-12-20

(Mann 1976; Alizadeh 2000). Surprisingly, however, there were subsets of
germinal
center B cell genes that were differentially expressed between BL and GCB.
[00112] NF-KB target genes were expressed at very low levels in BL as compared
to
the DLBCL subtypes. These genes are also known to be expressed at lower levels
in GCB than in ABC and PMBL (Rosenwald 2003b; Savage 2003; Feuerhake 2005;
Lam 2005). However, BL was found to express NF-KB target genes at levels even
lower than those of GCB. It is unclear at present whether this low expression
reflects differences in the malignant cells or in tumor-infiltrating immune
cells.
[00113] BL tumors expressed MHC class I genes at very low levels compared with
DLBCL tumors. Previous studies have documented the loss of MHC class I
molecules in some BL-derived cell lines (Voltz 1989), but the mechanism
underlying
this downmodulation is unclear at present.
[00114] The gene expression signatures that distinguish BL and DLBCL provide
insight into the nine BL-discrepant cases that were classified as BL by gene
expression but DLBCL by the panel of hematopathologists. The five BL-
discrepant
that were BCL2-negative were indistinguishable from the BL-concordant cases in
the
expression of all four gene expression signatures. Therefore, these cases bear
all
the hallmarks of BL but cannot be distinguished using current methodologies.
Interestingly, BL-discrepant cases that were BCL2-positive resembled the BL-
concordant cases with respect to three gene expression signatures, but had
lower
expression of the BL-high GC B cell signature. This phenotype was also
observed
in the BCL2-positive BL-concordant cases. Cases carrying dual translocations
t(8;14) and t(14;18) have been described previously as having a very
aggressive
course and a poor prognosis (Macpherson 1999). The data presented herein
confirm that CHOP-like regimens are not adequate to treat such patients.
47
CA 2897828 2019-12-20

[00115] The method of distinguishing BL and DLBCL disclosed herein provides a
more quantitative and reproducible diagnosis of BL than is afforded by current
methods based on morphology and immunophenotype. Such a method is clinically
important because BL is a curable malignancy. Previous studies have shown that
BL patients treated with CHOP-like chemotherapy regimens fare significantly
worse
than those who receive intensive regimens (Butler 1993; Magrath 1996; Smeland
2004). The results disclosed herein reveal that this also true for cases that
were
diagnosed as DLBCL pathologically but as BL based on gene expression. Correct
identification of these cases will greatly enhance the formulation of
treatment options
for these BL-discrepant patients.
[00116] The methods discussed herein for identifying and classifying lymphoma
subtypes have been used to identify and characterize cases of cyclin D1-
negative
MCL. Cyclin D1 overexpression has generally been considered essential to MCL
pathogenesis. In fact, the current WHO guidelines for diagnosing MCL rely on
morphologic examination and immunophenotyping, and require demonstration of
cyclin D1 overexpression and/or the t(11;14)(q13;q32) for confirmation.
Several
suspected cases of cyclin-D1 MCL have been identified in recent studies, but
these
cases have been controversial and difficult to substantiate. Most reported
cases of
cyclin Dl-negative MCL have been attributed to suboptimal immunostaining,
inadequate genetic or molecular analyses, or misdiagnosis.
[00117] Nevertheless, in a recent study of 99 lymphomas, seven cases were
identified that were morphologically consistent with MCL but lacked cyclin D1
expression as measured by quantitative RT-PCR and Lymphochip cDNA microarray
analyses (Rosenwald 2003). Other than cyclin D1, these cases exhibited
characteristic MCL gene expression signatures as determined by cDNA microarray
48
CA 2897828 2019-12-20

analysis, and were therefore classified as cyclin D1-negative MCL. One of
these
seven cases had the characteristic t(11;14)(q13;q32) as determined by FISH
analysis, and expressed cyclin D1 as determined by immunohistochemical
staining.
Thus, this case was determined to be a false negative and was reclassified as
cyclin
D1-positive MCL. Additional gene expression profiling analysis was performed
using
U133A/B microarrays, and the algorithm for diagnosing MCL was refined. Using
this
refined algorithm, four of the six remaining cyclin D1-negative cases were
determined to be unclassifiable B-cell lymphomas, and were thus excluded from
additional study. The two remaining cases and four newly identified cases were
used for the current studies.
[00118] The present disclosure confirms and extends previous findings
regarding the
identification of cyclin D1-negative MCL. Six cases of MCL have been confirmed
herein as negative for cyclin D1 mRNA expression by quantitative RT-PCR,
microarray analysis, and immunostaining. These cases also lack the
characteristic
IGH/CCND1 fusion by FISH analysis. Nonetheless, all six cases exhibit the
characteristic pathologic features of MCL and, more importantly, shared the
characteristic MCL gene expression profile by microarray analysis. Therefore,
these
cases are regarded as bone fide cases of cyclin D1-negative MCL. The existence
of
such cases sheds new light on the pathobiology of MCL and challenges the idea
that
cyclin D1 overexpression is essential to MCL pathogenesis. It is also shown
herein
that patients with cyclin Di-negative MCL have clinical and pathologic
features
similar to those with cyclin Dl-positive MCL. In particular, tumors in both
groups
have similar growth patterns and common cytological and immunohistochemical
features. Similar age and sex distribution, stage, serum LDH levels,
extranodal
49
CA 2897828 2019-12-20

sites, IPI scores, response to initial treatment, and overall survival are
observed in
the cyclin D1-positive and cyclin D1-negative groups.
[00119] Recent studies have reported 23 cyclin D1-negative cases among 151
cases of lymphoma with the morphological features of MCL based on
immunohistochemical staining (Yatabe 2000). Conventional cytogenetics was
performed on only three of these 23 cases, and all three were negative for the
t(11;14)(q13;q32). However, FISH or quantitative RT-PCR analysis was not
performed on any of these cases. Compared to the 128 cyclin D1-positive MCL
cases, the 23 cyclin D1-negative cases exhibited significantly better overall
survival.
Another recent study identified three cases of apparent cyclin D1-negative MCL
by
immunostains and compared them to 14 cases of typical cyclin D1-positive MCL
(Hashimoto 2002). This study suggested that cyclin D1-negative MCL is a more
indolent form of MCL. However, neither of these studies provided convincing
evidence that the cyclin D1-negative MCL disclosed therein were true cases of
MCL.
In fact, the Yatabe et al. study notes that some of the cases identified as
cyclin D1-
negative MCL might actually be marginal zone B-cell lymphomas or atypical
small
lymphocytic lymphomas (Yatabe 2000). The results presented herein represent
the
first demonstration of characteristic MCL gene expression signature in a set
of cyclin
D1-negative MCL cases. No significant difference in clinical features was
identified
between these cases and cyclin D1-positive MCL.
[00120] The pathogenic mechanisms involved in the development of the cyclin D1-
negative MCL are currently unknown. Since the oncogenic effect of
overexpressed
cyclin D1 is considered to be cell cycle dysregulation, other proteins
involved in cell
cycle control, especially the G1 to S phase transition, were examined. The D-
type
cyclins, D1, D2 and D3, are all positive promoters of cell cycle progression
from the
CA 2897828 2019-12-20

G1 to S phase. The D-type cyclins are similar in structure and biochemical
function
(Inaba 1992), but are expressed in a lineage-specific manner (Sherr 1994).
There is
considerable redundancy in the growth promoting function of the D-type
cyclins,
since only limited phenotypic consequences due to the absence of either cyclin
D1,
D2, or D3 are seen in gene knock-out mice (Sicinski 1995; Sicinski 1996;
Ciemerych
2002; Sicinska 2003). In non-neoplastic lymph nodes and tonsils, cyclin D2 is
found
mainly in interfollicular T-cells, whereas cyclin D3 is found in centroblasts
in
lymphoid follicles and in scattered B cells and T cells of the interfollicular
areas
(Teramoto 1999). However, cyclin D1 is not expressed in non-neoplastic T-cells
or
B-cells (Rosenberg 1991; Yang 1994). In low-grade B-cell malignancies,
overexpression of cyclin D2 mRNA was observed by Northern blot analysis in 29
of
34 CLL cases and in all seven LPC cases, but not in two cases of MCL (Delmer
1995). Cyclin D3 appears to be expressed more ubiquitously in B-cell
malignancies,
including FL, marginal zone lymphoma, and DLBCL (Ciemerych 2002), but is
usually
not expressed in lymphoid malignancies with either cyclin D1 or D2
overexpression
(Ott 1997; Doglioni 1998; Suzuki 1999). In the current study, overexpression
of
either cyclin D2 or D3 was observed in all six cases of cyclin Dl-negative
MCL,
indicating an important substitute role for these cyclins in the pathogenesis
of cyclin
D1-negative MCL. However, the mechanism of cyclin D2 or D3 up-regulation in
these cases remains unclear. No chromosomal translocations or gene
amplifications involving the cyclin D2 or D3 gene loci by FISH analysis were
identified in these cases. The findings herein are consistent with several
previous
studies which suggested that deregulation of cyclin D2 or D3 expression is
often due
to epigenetic mechanisms (Andreasson 1998; Bergsagel 2003; Pruneri 2003).
51
CA 2897828 2019-12-20

[00121]Dysregulation of other genes or factors important in cell cycle control
could
also play a role in the pathogenesis of such cases. These may include
dysregulation of p27kiPl, up-regulation of cyclin E, inactivation of the RB
gene,
deletion of the p16INK4a/p14ARF locus, as well as involvement of other genes.
The
protein regulates cellular progression from G1 into S phase by inhibiting the
cyclin E/CDK2 complex (Polyak 1994). Regulation of p27k1P1 occurs primarily
through posttranscriptional mechanisms, including sequestration by cyclin D1
or
cyclin D3 (Lin 2003; Quintanilla-Martinez 2003) or proteasomal degradation
(Chiarle
2000). In a prior study, expression of p27k1P1 as assessed by immunostains was
noted in only five of 40 cases of typical MCL, but was found in eight of ten
cases of
blastic MCL (Quintanilla-Martinez 1998). In the current study, down-regulation
of
protein expression was seen in all six cases, similar to that seen in typical
cyclin Dl-positive MCL.
[00122] The E-type cyclins, including cyclin El and E2, are also important in
the G1
phase of the cell cycle. When combined with CDK2, cyclin E promotes the hyper-
phosphorylation of RB protein, and thereby facilitates the entry of cells into
S phase
(Sherr 1996). However, none of the present cases were positive for cyclin E
expression, arguing against a role for cyclin E in the pathogenesis of cyclin
D1-
negative MCL. Inactivation of the RB tumor suppressor gene has been implicated
in
the development of various types of human malignancy. However, RB protein
expression was identified in all of our cases and the expression levels were
similar to
those seen in cyclin Dl-positive MCL. The present findings are consistent with
a
previous study concluding that RB protein appears to be normally regulated in
MCL
(Jares 1996). The present study also investigated whether deletions of the
tumor
suppressor genes p16INK42 and p 4ARF play a role in cyclin Dl-negative MCL.
52
CA 2897828 2019-12-20

p161"ma regulates the G1/S phase transition by forming binary complexes with
CDK
4 and 6, thereby preventing these subunits from association with D-type
cyclins
(Sherr 2002). Deletion of p16INK4a or cyclin D1 overexpression may therefore
promote the G1/S phase transition by the same mechanism. An important function
of p14ARF is to augment p53 function by antagonizing MDM2, and loss of p14ARF
function may contribute to the enhanced proliferation in tumor cells (Sherr
2002). As
has been shown previously, INK4a/ARF locus deletions occur in up to 21%
(18/85)
cases of MCL and are preferentially observed among the more proliferative
cases
(Rosenwald 2003). However, deletion of the INK4a/ARF locus was not identified
in
any of the six cases, arguing against a role for p16INK4a/p14ARF in the
pathogenesis of
cyclin Dl-negative MCL.
[00123] The classification of a lymphoproliferative disorder in accordance
with
embodiments of the present invention may be used in combination with any other
effective classification feature or set of features. For example, a disorder
may be
classified by a method of the present invention in conjunction with WHO
suggested
guidelines, morphological properties, histochemical properties, chromosomal
structure, genetic mutation, cellular proliferation rates, immunoreactivity,
clinical
presentation, and/or response to chemical, biological, or other agents.
Embodiments of the present invention may be used in lieu of or in conjunction
with
other methods for lymphoma diagnosis, such as immunohistochemistry, flow
cytometry, FISH for translocations, or viral diagnostics.
[00124] Accurate determination of lymphoma type in a subject allows for better
selection and application of therapeutic methods. Knowledge about the exact
lymphoma affecting a subject allows a clinician to select therapies or
treatments that
are most appropriate and useful for that subject, while avoiding therapies
that are
53
CA 2897828 2019-12-20

nonproductive or even counterproductive. For example, CNS prophylaxis may be
useful for treating BL but not DLBCL, CHOP treatment may be useful for
treating
DLBCL but not blastic MCL (Fisher 1993; Khouri 1998), and subjects with
follicular
lymphoma frequently receive treatment while subjects with follicular
hyperplasia do
not. In each of these situations, the lymphoma types or subtypes in question
can be
difficult to distinguish using prior art diagnostic methods. The diagnostic
and
identification methods of the present invention allow for more precise
delineation
between these lymphomas, which simplifies the decision of whether to pursue a
particular therapeutic option. Likewise, the survival prediction methods
disclosed in
the present invention also allow for better selection of therapeutic options.
A subject
with a very low survival predictor score (i.e., very good prognosis) may not
receive
treatment, but may instead be subjected to periodic check-ups and diligent
observation. As survival predictor scores increase (i.e., prognosis gets
worse),
subjects may receive more intensive treatments. Those subjects with the
highest
survival predictor scores (i.e., very poor prognosis) may receive experimental
treatments or treatments with novel agents. Accurate survival prediction using
the
methods disclosed herein provides an improved tool for selecting treatment
options
and for predicting the likely clinical outcome of those options.
[00125] Any effective method of quantifying the expression of at least one
gene,
gene set, or group of gene sets may be used to acquire gene expression data
for
use in embodiments of the present invention. For example, gene expression data
may be measured or estimated using one or more microarrays. The microarrays
may be of any effective type, including but not limited to nucleic acid based
or
antibody based. Gene expression may also be measured by a variety of other
techniques, including but not limited to PCR, quantitative RT-PCR, real-time
PCR,
54
CA 2897828 2019-12-20

RNA amplification, in situ hybridization, innmunohistochemistry,
immunocytochemistry, FACS, serial analysis of gene expression (SAGE)
(Velculescu 1995), Northern blot hybridization, or western blot hybridization.
[00126] Nucleic acid microarrays generally comprise nucleic acid probes
derived
from individual genes and placed in an ordered array on a support. This
support
may be, for example, a glass slide, a nylon membrane, or a silicon wafer. Gene
expression patterns in a sample are obtained by hybridizing the microarray
with the
gene expression product from the sample. This gene expression product may be,
for example, total cellular mRNA, rRNA, or cDNA obtained by reverse
transcription
of total cellular mRNA. The gene expression product from a sample is labeled
with a
radioactive, fluorescent, or other label to allow for detection. Following
hybridization,
the microarray is washed, and hybridization of gene expression product to each
nucleic acid probe on the microarray is detected and quantified using a
detection
device such as a phosphorimager or scanning confocal microscope.
[00127] There are two broad classes of microarrays: cDNA and oligonucleotide
arrays. cDNA arrays consist of hundreds or thousands of cDNA probes
immobilized
on a solid support. These cDNA probes are usually 100 nucleotides or greater
in
size. There are two commonly used designs for cDNA arrays. The first is the
nitrocellulose filter array, which is generally prepared by robotic spotting
of purified
DNA fragments or lysates of bacteria containing cDNA clones onto a
nitrocellulose
filter (Southern 1992; Southern 1994; Gress 1996; Pietu 1996). The other
commonly used cDNA arrays is fabricated by robotic spotting of PCR fragments
from cDNA clones onto glass microscope slides (Schena 1995; DeRisi 1996;
Schena 1996; Shalon 1996; DeRisi 1997; Heller 1997; Lashkari 1997). These cDNA
microarrays are simultaneously hybridized with two fluorescent cDNA probes,
each
CA 2897828 2019-12-20

labeled with a different fluorescent dye (typically Cy3 or Cy5). In this
format, the
relative mRNA expression in two samples is directly compared for each gene on
the
microarray. Oligonucleotide arrays differ from cDNA arrays in that the probes
are
20- to 25-mer oligonucleotides. Oligonucleotide arrays are generally produced
by in
situ oligonucleotide synthesis in conjunction with photolithographic masking
techniques (Pease 1994; Lipshutz 1995; Chee 1996; Lockhart 1996; Wodicka
1997). The solid support for oligonucleotide arrays is typically a glass or
silicon
surface.
[00128] Methods and techniques applicable to array synthesis and use have been
described in, for example, U.S. Patent Nos. 5,143,854 (Pirrung), 5,242,974
(Holmes), 5,252,743 (Barrett), 5,324,633 (Fodor), 5,384,261 (Winkler),
5,424,186
(Fodor), 5,445,934 (Fodor), 5,451,683 (Barrett), 5,482,867 (Barrett),
5,491,074
(Aldwin), 5,527,681 (Holmes), 5,550,215 (Holmes), 5,571,639 (Hubbell),
5,578,832
(Trulson), 5,593,839 (Hubbell), 5,599,695 (Pease), 5,624,711 (Sundberg),
5,631,734
(Stern), 5,795,716 (Chee), 5,831,070 (Pease), 5,837,832 (Chee), 5,856,101
(Hubbell), 5,858,659 (Sapo'sky), 5,936,324 (Montagu), 5,968,740 (Fodor),
5,974,164
(Ghee), 5,981,185 (Matson), 5,981,956 (Stern), 6,025,601 (Trulson), 6,033,860
(Lockhart), 6,040,193 (Winkler), 6,090,555 (Fiekowsky), and 6,410,229
(Lockhart),
and U.S. Patent Application Publication No. 20030104411 (Fodor).
[00129] Microarrays may generally be produced using a variety of techniques,
such
as mechanical or light directed synthesis methods that incorporate a
combination of
photolithographic methods and solid phase synthesis methods. Techniques for
the
synthesis of microarrays using mechanical synthesis methods are described in,
for
example, U.S. Patent Nos. 5,384,261 (Winkler) and 6,040,193 (Winkler).
Although a
planar array surface is preferred, the microarray may be fabricated on a
surface of
56
CA 2897828 2019-12-20

virtually any shape, or even on a multiplicity of surfaces. Microarrays may be
nucleic
acids on beads, gels, polymeric surfaces, fibers such as fiber optics, glass
or any
other appropriate substrate. See, for example, U.S. Patent Nos. 5,708,153
(Dower);
5,770,358 (Dower); 5,789,162 (Dower); 5,800,992 (Fodor); and 6,040,193
(Winkler).
[00130] Microarrays may be packaged in such a manner as to allow for
diagnostic
use, or they can be an all-inclusive device. See, for example, U.S. Patent
Nos.
5,856,174 (Lipshutz) and 5,922,591 (Anderson).
[00131] Microarrays directed to a variety of purposes are commercially
available
from Affymetrix (Affymetrix, Santa Clara, CA). For instance, these microarrays
may
be used for genotyping and gene expression monitoring for a variety of
eukaryotic
and prokaryotic species.
[00132] The following examples are provided to better illustrate the claimed
invention
and are not to be interpreted as limiting the scope of the invention. To the
extent
that specific materials are mentioned, it is merely for purposes of
illustration and is
not intended to limit the invention. One skilled in the art may develop
equivalent
means or reactants without the exercise of inventive capacity and without
departing
from the scope of the invention. It will be understood that many variations
can be
made in the procedures herein described while still remaining within the
bounds of
the present invention. It is the intention of the inventors that such
variations are
included within the scope of the invention.
Examples
Example 1: Collection and analysis of gene expression data using Affymetrix
U133A
and U133B microarrays:
[00133] 568 cell samples representing various forms of human lymphoid
malignancies were obtained by biopsy using known methods described in the
57
CA 2897828 2019-12-20

literature. The samples were reviewed by a panel of hematopathologists and
classified into the following lymphoma types based on current diagnostic
criteria:
231 diffuse large B cell lymphomas (DLBCL)
191 follicular lymphomas (FL)
26 Burkitt lymphomas (BL)
21 mantle cell lymphoma (MCL)
18 follicular hyperplasias (FH)
17 small cell lymphocytic lymphomas (SLL)
16 mucosa-associated lymphoid tissue lymphomas (MALT)
13 splenic lymphomas (Splenic)
cyclin-D1 negative lymphomas with MCL morphology (CD1negMCL)
9 multiple myeloma (Mult_Myeloma)
6 lymphoplasmacytic lymphomas (LPC)
4 post-transplant lymphoproliferative disorders (PTLD)
3 lymphoblastic lymphomas (Lymbl)
3 nodal marginal zone lymphomas (NMZ)
The 231 DLBCL samples were subdivided into the following lymphoma types based
on gene expression (see below):
88 germinal center B cell-like (GCB)
78 activated B cell-like (ABC)
33 primary mediastinal B cell lymphoma (PMBL)
32 samples for which the subtype could not be determined (UC_DLBCL)
The 16 MALT samples were subdivided into the following four group based on
tumor
origin:
9 from the gastric region (MALT_gastric)
58
CA 2897828 2019-12-20

I from the salivary gland (MALT_salivary)
1 from the lung (MALT _lung)
1 from the tonsil (MALT_tonsil)
4 of unknown origin (MALT_unk)
[00134] Each of the 568 cell samples was given a unique sample ID number
consisting of the lymphoma type followed by a unique numerical identifier. For
example, "ABC_304" refers to an ABC DLBCL sample numbered 304. Cells were
purified and RNA was isolated from the purified cells according to known
methods
described in the literature.
[00135] Aliquots of RNA from each sample were applied to Affymetrix U133A
and Affymetrix U133B microarrays according to standard Affymetrix protocol.
The U133A and U133B microarrays are divided into probe sets, with each
probe set consisting of up to 69 oligonucleotide probes 25 nucleotides in
length. Each probe set represents a distinct human gene. Information
pertaining to these microarrays is available a the Affymetrix company web
site.
Each microarray was scanned using an Affymetrix scanner, which records
signal intensity for every probe on the microarray. This information can be
transformed into summary signal values for each probe set using a number of
different algorithms, including MAS 5.0, D-chip (Li 2001), or Bioconductor's
RMA algorithms (Irizarry 2003). The images produced by the scanner were
evaluated by Affymetrix MAS 5.0 software and stored as tables in .txt format.
Since each sample was scanned on both microarrays, there are two .txt files
for each sample. Each .txt file was given a unique name consisting of the
table number, sample ID number (discussed above), and a letter denoting the
59
CA 2897828 2019-12-20

microarray used. For example, Table_0588_ABC_304_A.txt is the .txt file for
Table 588, which contains data for sample ID number ABC_304 from the
U133A array. The data for each sample tested is contained in Tables 3-1138.
[00136] The signal value for each probe on the U133A and U133B microarrays was
normalized to a target value of 500, and the base-2 log of the normalized
values was
used for the following analyses. Log-signal values for each probe set are
presented
in Tables 1139-1706, contained in files with the title format
"Table_No._NAME Jog_signal.txt," where NAME refers to the sample ID number
(e.g., ABC_304). The first column provides the UNIQID for the probe set, while
the
second column provides the log-signal value.
[00137] Log-signal files were statistically analyzed using S+ software and the
S+
subtype predictor script contained in the file entitled
"Subtype_Predictor.txt," located
in the computer program listing appendix contained on CD number 22 of 22.
Although the log-signal values were analyzed using S+ software and the above
algorithm, any effective software/algorithm combination may be used. Tables
1707-
1721 provide descriptive statistical characteristics for each of the lymphoma
types
tested except for CD1negMCL, non-gastric MALT, and UC_DLBCL. Table 1722
provides statistical characteristics for all MALT samples combined, while
Table 1723
does likewise for all DLBCL samples.
[00138] The files containing Tables 1707-1723 have the title format
"Table_No._TYPE_Stats.txt," where TYPE refers to the lymphoma type. Each row
of these tables represents a particular probe set. The first column of each
table
provides the UNIQID for the probe set, while the second column provides the
average log-signal for the probe set over all samples of a particular lymphoma
type.
The third column provides the log-fold change in expression of the probe set
CA 2897828 2019-12-20

between the lymphoma type in question and a second lymphoma type. For
example, if logfold.ABC.vs.GCB is -0.21 for gene X, expression of gene X in
the
ABC samples is, on average, 0.86 (i.e., 2-021) times greater than expression
of gene
X in the GCB samples. The fourth column provides a two-sided P-value derived
from a t-test of the log signals of the two lymphoma types compared in column
three.
If, for example, P.value.ABC.vs.GCB was 0.00001 for gene X, this would
indicate
that the observed difference in expression of gene X between ABC and GCB would
only occur approximately one time in 100,000 if there was no actual difference
in
gene X expression between the two lymphoma types. The remainder of the
columns can be read as pairs that repeat the pattern of columns three and
four,
presenting the log-fold change and P-value of the difference in expression of
the
probe set for the lymphoma type in question versus all other lymphoma types
being
tested. Tables 1710, 1715, and 1723 (corresponding to FL, MCL, and DLBCL,
respectively) contain two additional columns entitled "TYPE_Cox_coefficient"
and
"TYPE_Cox_P_value." The content of these columns is discussed in the following
examples.
Example 2: Collection of gene expression data using the novel Lymph Dx
microarray:
[00139] The novel Lymph Dx microarray contains cDNA probes corresponding to
approximately 2,734 genes. 174 of these are "housekeeping" genes present for
quality control, since they represent genes that are most variably expressed
across
all lymphoma samples. Other genes represented on the microarray were selected
for their utility in identifying particular lymphoma samples and predicting
survival in
those samples. The genes represented on the Lymph Dx microarray can be divided
into four broad categories: 1,101 lymphoma predictor genes identified
previously
61
CA 2897828 2019-12-20

using the Affymetrix U133 microarray, 171 outcome predictor genes identified
using
the Affymetrix U133 microarray, 167 genes not found on the Affymetrix U133
microarray but represented on the Lymphochip microarray (Alizadeh 1999), and
1,121 named genes. The types of genes making up each of these broad categories
are summarized in Table 1724, below, while the specific genes represented on
the
Lymph Dx microarray are listed in Table 2, contained in the file
"Table_0002_LymphDx_Probe_List.txt."
Table 1724
Gene type Number of
genes
Lymphoma predictor genes 1101
Subtype specific 763
Lymph node signature 178
Proliferation signature 160
Outcome predictor genes 171
DLBCL 79
FL 81
MCL 11
New genes not on U133 167
Lymphochip lymphoma predictor genes 84
EBV and HHV8 viral genes 18
BCL-2/cyclin D1/INK4a specialty probes 14
Named genes missing from U133 51
Named genes 1121
Protein kinase 440
Interleukin 35
Interleukin receptor 29
Chemokine 51
Chemokine receptor 29
TNF family 26
TNF receptor family 51
Adhesion 45
Surface marker 264
Oncogene/tumor suppressor 49
Apoptosis 46
Drug target 10
Regulatory 46
[00140] Cell samples representing various forms of human lymphoid malignancy
were obtained by biopsy using known methods described in the literature. These
634 biopsy samples were reviewed by a panel of hematopathologists and
classified
into the following lymphoma types based on current diagnostic criteria:
62
CA 2897828 2019-12-20

201 diffuse large B-cell lymphomas (DLBCL)
191 follicular lymphomas (FL)
60 Burkitt lymphomas (BL)
21 mantle cell lymphomas (MCL)
30 primary mediastinal B cell lymphoma (PMBL)
18 follicular hyperplasias (FH)
18 small cell lymphocytic lymphomas (SLL)
17 mucosa-associated lymphoid tissue lymphomas (MALT), including 9
gastric MALTs (GMALT)
16 chronic lymphocytic leukemias (CLL)
13 splenic lymphomas (SPL)
11 lymphoplasmacytic lymphomas (LPC)
11 transformed DLBCL (trDLBCL) (DLBCL that arose from an antecedent FL)
cyclin D1 negative lymphomas with MCL morphology (CD1N)
6 peripheral T-cell lymphoma (PTCL)
4 post-transplant lymphoproliferative disorders (PTLD)
4 nodal marginal zone lymphomas (NMZ)
3 lymphoblastic lymphomas (LBL)
[00141] Each of the 634 samples was given a unique sample ID number consisting
of the lymphoma type followed by a unique numerical identifier. For example,
"BL_2032_52748" refers to a Burkitt lymphoma sample with the numerical
identifier
2032_52748. Cells were purified and RNA was isolated from the purified cells
according to known methods described in the literature.
[00142] Aliquots of purified RNA from each sample were applied to the Lymph Dx
microarrays according to standard Affymetrix microarray protocol. Each
microarray
63
CA 2897828 2019-12-20

was scanned on an Affymetrix scanner. This scanner produced an image of the
microarray, which was then evaluated by Affymetrix MAS 5.0 software. This
information was stored in tables in .txt format. Each of these .txt files was
given a
unique name consisting of the table number, the sample ID number (discussed
above), and the UNIQID for identifying the array data in the National Cancer
Institute
Database. For example, Table_1725_BL_2032_52748.txt is the .txt file for Table
1725, which contains data for sample ID number BL_2032. The data for each
sample analyzed is contained in Tables 1725-2358. The signal intensity for
each
probe on the microarray can be transformed into summary signal values for each
probe set through a number of different algorithms, including but not limited
to MAS
5.0, D-chip (Li 2001), or Bioconductor's RMA algorithms (Irizarry 2003).
Example 3: Development of a first FL survival predictor using gene expression
data
from Affvmetrix U133A and U133B microarrays:
[00143] An analytical method entitled Survival Signature Analysis was
developed to
create survival prediction models for lymphoma. This method is summarized in
Figure 2. The key feature of this method is the identification of gene
expression
signatures. Survival Signature Analysis begins by identifying genes whose
expression patterns are statistically associated with survival. A hierarchical
clustering algorithm is then used to identify subsets of these genes with
correlated
expression patterns across the lymphoma samples. These subsets are
operationally defined as "survival-associated signatures." Evaluating a
limited
number of survival-associated signatures mitigates the multiple comparison
problems that are inherent in the use of large-scale gene expression data sets
to
create statistical models of survival (Ransohoff 2004).
64
CA 2897828 2019-12-20

[00144] FL samples were divided into two equivalent groups: a training set (95
samples) for developing the survival prediction model, and a validation set
(96
samples) for evaluating the reproducibility of the model. The overall survival
of this
cohort is depicted in Figure 3. The median age at diagnosis was 51 years
(ranging
from 23 to 81 years), and the patients had a median follow-up of 6.6 years
(8.1
years for survivors, with a range of <1 to 28.2 years). Gene expression data
from
Affymetrix U133A and U133B microarrays was obtained for each sample. Within
the
training set, a Cox proportional hazards model was used to identify "survival
predictor" genes, which were genes whose expression levels were associated
with
long survival (good prognosis genes) or short survival (poor prognosis genes).
A
hierarchical clustering algorithm (Eisen 1998) was used to identify gene
expression
signatures within the good and poor prognosis genes according to their
expression
pattern across all samples. Ten gene expression signatures were observed
within
either the good prognosis or poor prognosis gene sets (Figure 4). The
expression
level of every component gene in each of these ten gene expression signatures
was
averaged to create a gene expression signature value.
[00145] To create a multivariate model of survival, different combinations of
the
ten gene expression signature values were generated and evaluated for their
ability to predict survival within the training set. Among models consisting
of
two signatures, an exceptionally strong statistical synergy was observed
between one signature from the good prognosis group and one signature from
the poor prognosis group. These signatures were deemed "immune
response-1" and "immune response-2," respectively; based on the biological
function of certain genes within each signature. The immune response-1 gene
expression signature included genes encoding T cell markers (e.g., CD7,
CA 2897828 2019-12-20

CD8B1, ITK, LEF1, STAT4) and genes that are highly expressed in
macrophages (e.g., ACTN1, TNFSF13B). The immune response-1 signature
is not merely a surrogate for the number of T cells in the FL biopsy sample
because many other standard T cell genes (e.g., CD2, CD4, LAT, TRIM,
SH2D1A) were not associated with survival. The immune response-2 gene
expression signature included genes known to be preferentially expressed in
macrophages and/or dendritic cells (e.g., TLR5, FCGR1A, SEPT10, LGMN,
C3AR1). Table 2359 lists the genes that were used to generate the gene
expression signature values for the immune response-1 and immune
response-2 signatures. The Unigene ID Build database referenced in the
following tables is available at the National Center for Biotechnology
Information (NCBI) web site.
Table 2359
Signature UNIQID Unigene ID Build 167 Gene symbol
Immune response-1 1095985 83883 TMEPAI
Immune response-1 1096579 117339 HCST
Immune response-1 1097255 380144
Immune response-1 1097307 379754 LOC340061
Immune response-1 1097329 528675 TEAD1
Immune response-1 1097561 19221 C20orf112
Immune response-1 1098152 377588 KIAA1450
Immune response-1 1098405 362807 IL7R
Immune response-1 1098548 436639 NFIC
Immune response-1 1098893 43577 ATP8B2
Immune response-1 1099053 376041
Immune response-1 1100871 48353
Immune response-1 1101004 2969 SKI
Immune response-1 1103303 49605 C9or152
Immune response-1 1107713 171806
Immune response-1 1115194 270737 TNFSF13B
Immune response-1 1119251 433941 SEPW1
Immune response-1 1119838 469951 GNAQ
Immune response-1 1119924 32309 INPP1
Immune response-1 1120196 173802 TBC1D4
Immune response-1 1120267 256278 TNFRSF1B
Immune response-1 1121313 290432 HOXB2
Immune response-1 1121406 NA TNFSF12
Immune response-1 1121720 80642 STAT4
Immune response-1 1122956 113987 LGALS2
Immune response-1 1123038 119000 ACTN1
66
CA 2897828 2019-12-20

Immune response-1 1123092 437191 PTRF
Immune response-1 1123875 428 FLT3LG
Immune response-1 1124760 419149 JAM3
Immune response-1 1128356 415792 C1RL
Immune response-1 1128395 7188 SEMA4C
Immune response-1 1132104 173802 TBC1D4
Immune response-1 1133408 12802 DDEF2
Immune response-1 1134069 405667 CD8B1
Immune response-1 1134751 106185 RALGDS
Immune response-1 1134945 81897 KIAA1128
Immune response-1 1135743 299558 TNFRSF25
Immune response-1 1135968 119000 ACTN1
Immune response-1 1136048 299558 TNFRSF25
Immune response-1 1136087 211576 ITK
Immune response-1 1137137 195464 FLNA
Immune response-1 1137289 36972 CD7
Immune response-1 1137534 36972 CD7
Immune response-1 1139339 47099 GALNT12
Immune response-1 1139461 14770 BIN2
Immune response-1 1140391 44865 LEF1
Immune response-1 1140524 10784 C6orf37
Immune response-1 1140759 298530 RAB27A
Immune response-2 1118755 127826 EPOR
Immune response-2 1118966 19196 L0051619
Immune response-2 1121053 1690 FGFBP1
Immune response-2 1121267 334629 SLN
Immune response-2 1121331 8980 TESK2
Immune response-2 1121766 396566 MPP3
Immune response-2 1121852 421391 LECT1
Immune response-2 1122624 126378 ABCG4
Immune response-2 1122679 232770 ALOXE3
Immune response-2 1122770 66578 CRHR2
Immune response-2 1123767 1309 CD1A
Immune response-2 1123841 389 ADH7
Immune response-2 1126097 498015
Immune response-2 1126380 159408
Immune response-2 1126628 254321 CTNNA1
Immune response-2 1126836 414410 NEK1
Immune response-2 1127277 121494 SPAM1
Immune response-2 1127519 NA
Immune response-2 1127648 285050
Immune response-2 1128483 444359 SEMA4G
Immune response-2 1128818 115830 HS3ST2
Immune response-2 1129012 95497 SLC2A9
Immune response-2 1129582 272236 C21orf77
Immune response-2 1129658 _ 58356 PGLYRP4
Immune response-2 1129705 289368 ADAM19
Immune response-2 1129867 283963 G6PC2
Immune response-2 1130003 432799
Immune response-2 1130388 19196 L0051619
Immune response-2 1131837 156114 PTPNS1
Immune response-2 1133843 6682 SLC7A11
_
Immune response-2 1133949 502092 PSG9
Immune response-2 1134447 417628 CRHR1
Immune response-2 1135117 512646 PSG6
Immune response-2 1136017 1645 CYP4A11
Immune response-2 1137478 315235 ALDOB
Immune response-2 1137745 26776 NTRK3
67
CA 2897828 2019-12-20

Immune response-2 1137768 479985
Immune response-2 1138476 351874 HLA-DOA
Immune response-2 1138529 407604 CRSP2
Immune response-2 1138601 149473 PRSS7
Immune response-2 1139862 251383 CHST4
Immune response-2 1140189 287369 IL22
Immune response-2 1140389 22116 CDC14B
[00146]Although the immune response-1 and immune response-2 gene expression
signatures taken individually were not ideal predictors of survival, the
binary model
formed by combining the two was more predictive of survival in the training
set than
any other binary model (p<0.001). Using this binary model as an anchor, other
signatures were added to the model using a step up procedure (Drapner 1966).
Of
the remaining eight signatures, only one signature contributed significantly
to the
model in the training set (p<0.01), resulting in a three-variable model for
survival.
This model was associated with survival in a highly statistically significant
fashion in
both the training (p<0.001) and validation sets (p=0.003). However, only the
immune response-1 and immune response-2 gene expression signatures
contributed to the predictive power of the model in both the training set and
the
validation set. The predictive power of each of these signatures is summarized
in
Table 2360.
Table 2360
Gene expression Contribution of Relative
risk of death Effect of increased
signature signature to model in among patients in
expression on
validation set (p- validation set (95% survival
value)
Immune response-1 <0.001 0.15 (0.05-0.46) Favorable
Immune response-2 <0.001 9.35 (3.02-28.9) Poor
Based on this information, the third signature was removed from the model and
the
two-signature model was used to generate a survival predictor score using the
following equation:
68
CA 2897828 2019-12-20

Survival predictor score = [(2.71*immune response-2 gene expression
signature value)] - [(2.36 x immune response-1 gene expression
signature value)].
A higher survival predictor score was associated with worse outcome. The two-
signature model was associated with survival in a statistically significant
fashion in
both the training set (p<0.001) and the validation set (p<0.001), which
demonstrated
that the model was reproducible. For the 187 FL samples with available
clinical
data, the survival predictor score had a mean of 1.6 and a standard deviation
of
0.894, with each unit increase in the predictor score corresponding to a 2.5
fold
increase in the relative risk of death. Data for all 191 samples is shown in
Table
2361.
Table 2361
Sample Set Length of Status Immune Immune
Survival
ID # follow-up at response-1 response-2 predictor
(years) follow-up signature value signature value score
FL 1073 Training 7.68 Dead 9.20 8.67 1.77
FL_1074 Training 4.52 Dead 9.10 8.57 1.74
FL 1075 Validation 4.52 Dead 8.97 8.69 2.38
FL_1076 Training 3.22 Dead 9.20 8.55 1.44
FL 1077 Training 7.06 Alive 9.80 8.46 -0.20
FL_1078 Training 4.95 Alive , 9.32 8.23 0.30
FL 1080 Training 6.05 Alive 9.45 8.94 1.93
FL_1081 Validation 6.61 Alive 9.00 8.22 1.05
FL_1083 Training 10.01 Alive 9.82 8.72 0.47
FL_1085 Validation 8.84 Alive 9.31 8.58 1.29
FL 1086 Validation 1.98 Dead 9.49 9.09 2.22
FL_1087 Training 8.19 Alive 9.98 9.27 1.57
FL_1088 Validation 5.30 Alive 9.22 8.47 1.20
FL 1089 Training 10.72 Alive 9.42 8.35 0.40
_ .
FL_1090 Validation . 10.20 Alive 9.27 8.37 0.82
FL 1097 Validation 8.79 Dead 9.87 8.92 0.87
FL_1098 Validation 5.34 Dead 9.33 - 8.81 1.87
FL_1099 Training 7.65 Alive , 9.73 9.04 1.54
FL_1102 Validation _ 13.20 Dead 9.45 8.89 1.79
FL_1104 Training . 8.42 Dead _ 9.30 8.27 0.48
FL_1106 Validation 7.94 Alive 9.13 9.19 3.36
FL_1107 Training 5.01 , Dead , 9.41 9.32 3.07
FL 1183 Training 11.56 Dead 9.31 8.53 1.16
FL_1184 Training 6.93 Dead 9.66 8.83 1.13
FL_1185 Validation 7.02 Dead 9.23 9.09 2.86
Fl_1186 Training 1.34 Dead 9.01 8.84 2.68
FL 1416 Validation 6.21 Alive 9.50 8.67 1.08
FL_1417 Training 2.40 Dead 8.47 8.39 2.73
69
CA 2897828 2019-12-20

FL 1418 Validation 3.59 Alive 8.94 8.42 1.72
FL_1419 Training 3.85 Alive 9.82 8.56 0.03
FL 1422 Training 5.72 _ Alive 9.46 8.49 0.68
FL 1425 Validation 4.26 Alive 8.93 8.50 1.98
FL_1426 Training 7.32 Alive 9.08 8.26 0.97
FL 1427 Training 5.22 Alive 8.57 8.28 2.22
FL 1428 Validation 5.41 Dead 9.22 8.44 1.10
FL 1432 Training 3.66 Alive 9.22 8.95 2.51
FL 1436 Training 9.08 Dead 9.48 8.63 1.02
FL_1440 Training 7.85 , Alive 9.07 8.35 1.22
FL_1445 Training 9.24 _ Dead 8.67 8.66 3.01
FL_1450 Validation 0.65 Dead , 9.83 9.99 3.86
FL_1472 Validation 16.72 Alive 8.85 8.49 2.10
, FL_1473 Training 15.07 Alive 9.75 8.50 0.02
FL 1474 Validation 2.75 Dead 9.34 9.10 2.62
FL_1476 Validation 4.08 Dead 9.51 8.87 1.60
FL 1477 _ Training 0.59 Dead , 9.64 9.06 1.83
FL 1478 , Training 12.47 Dead 9.60 8.87 1.39
FL 1479 Training 2.29 Dead 8.71 9.07 4.01
FL_1480 Training 16.29 Alive 9.40 8.67 1.30
_FL 1579 Training_ 8.22 _ Dead 8.81 8.44
2.10
FL_1580 Training 19.30 Alive 9.58 8.52 0.49
FL_1581 Training 9.52 Dead 9.08 9.02 3.00
, FL 1582 Validation 1.30 Dead _ 8.40 8.18 2.36
FL 1583 Training 15.26 Dead _ 9.47 8.79 1.48
_FL_1584 Training 15.73 Dead _ 9.44 8.55 0.89
FL_1585 Validation 0.01 Alive _ 8.96 8.53 1.96
FL 1586 Validation 3.11 Alive 9.38 8.55 1.03
_ FL 1588 Training 0.49 Dead _ 9.52 9.06 2.08
FL_1589 Training 3.15 Alive _ 9.72 8.74 0.72
FL_1591 Training 11.22 Alive - 9.49 8.62 0.97
FL_1594 Validation 11.19 Alive
_ 9.25 8.59 1.47
FL_1595 Training 8.03 Alive _ 9.75 9.60 3.01
FL_1598 Validation 2.80 Dead 8.81 8.33 1.79
FL 1599 Validation 6.17 Alive - .. 9.48 8.65 1.06
FL_1603 Training 5.17 Dead 9.66 9.75 3.63
FL_1604 Training 3.98 Dead , 9.24 8.86 2.20
FL 1606 Validation 4.22 Dead 9.45 9.18 2.57
_
FL_1607 Validation 8.12 Alive . 9.40 8.60 1.13
1
FL_1608 Validation 9.70 Alive _ 8.92 8.41 1.72
FL_1610 Validation 2.05 Dead , 9.33 9.35 3.32
FL_1611 Validation 10.15 Alive _ 9.42 8.69 1.31
FL_1616 . Training 2.36 Dead 9.38 8.82 1.78
_
FL 1617 Validation 7.85 Alive _ 8.96 8.49 1.87
FL_1619 Validation 9.24 Dead 9.43 8.56 0.94
FL -1620 Validation 9.36 Dead 9.14 8.35 1.04
,
FL_1622 Training 14.01 Alive _ 9.23 8.53 1.33
FL 1623 Training 9.72 Alive 9.67 8.93 1.38
_ - r
FL_1624 Validation 3.98 Dead 9.05 8.50 1.70
FL_1625 _ Validation 11.16 Alive -
8.98 8.47 1.75
FL 1626 Validation 6.47 Dead ,_ 8.59 8.14 1.76
FL 1628 Validation 0.82 Dead - 9.80 8.72 0.51
_ -
_ FL_1637 Validation 18.81 Alive 9.95 9.58 2.48
_
FL 1638 Validation 4.06 Alive 9.13 8.88 2.51
_FL_1639 Training 4.75 Alive 9.53 8.89 1.62
_ FL_1643 Training 0.77 Dead - 9.73 9.06 1.58
FL -1644 Validation 3.84 Alive 9.55 8.68 0.98
_
_FL 1645 Training 3.56 Alive - 9.49 8.70 1.18
CA 2897828 2019-12-20

FL_1646 Training 1.97 Dead 9.25 8.61 1.50
FL 1647 Training 1.22 Dead 9.12 8.89 2.55
FL_1648 Training 11.01 Alive 9.13 8.12 0.46
FL_1652 Training 3.72 Dead 9.50 9.14 2.35
FL 1654 Validation 0.30 Dead 8.74 8.28 1.82
FL 1655 Training 8.45 Alive 9.51 8.85 1.53
FL 1656 Validation 9.36 Alive 9.06 8.58 1.87
FL_1657 Training 10.09 Alive 9.53 8.46 0.44
FL_1660 Training 2.32 Alive 8.81 8.38 1.91
FL_1661 Validation 1.48 Alive 9.86 8.90 0.85
FL_1662 Validation 0.74 Dead 9.57 9.15 2.21
FL 1664 Validation 4.53 Dead 9.34 8.62 1.31
FL_1669 Training 4.40 Dead 8.87 8.58 2.30
FL_1670 Training 1.88 Alive 9.64 9.45 2.86
FL 1675 Training 4.57 - Alive 9.36 8.46 0.84
FL_1681 Validation 4.23 Alive 9.52 8.63 0.91 _
FL_1683 Validation 4.03 Dead 9.95 9.10 1.19
FL_1684 Training 2.88 Dead 9.53 8.73 1.18
FL 1716 Validation 9.69 Alive 8.95 8.35 , 1.50
FL_1717 Validation 2.01 Dead 9.35 8.88 1.98
FL_1718 Training 10.35 Alive 9.23 8.13 0.26
FL 1719 Validation 7.70 Dead 9.13 8.50 1.49
FL_1720 Training 3.91 Dead 8.78 8.88 3.33
FL_1729 Training 8.06 Alive 9.35 8.65 1.39
FL 1732 Validation 0.71 Dead 7.81 8.59 4.86
FL_1761 Validation 10.83 Alive 9.31 8.55 1.22
FL_1764 Training 0.42 Dead 9.25 8.87 2.21
FL_1768 Training 13.04 , Alive 9.42 8.47 0.72
FL 1771 Training 9.26 Dead 9.09 8.67 2.06
FL 1772 Validation 13.64 Dead 9.49 8.49 0.61
FL_1788 Training 1.00 Dead 9.09 9.13 3.29
7 FL_1790 Training 1.42 Alive 9.85 9.40 2.22
FL_1792 Validation 2.01 Dead 9.33 8.72 1.61
,
FL_1795 Training 0.71 Dead 10.19 9.27 1.08
FL _1797 Validation 7.17 Alive 9.34 8.92 2.14
_
FL -1799 Training 14.18 Alive 9.32 8.63 1.38
_
FL_1810 Validation 9.91 Alive 8.66 8.41 2.35
FL_1811 Validation 3.04 Alive 9.38 8.27 0.29
FL 1825 Training 2.98 Alive 9.46 9.07 2.25
FL_1827 Training 3.66 Alive 9.80 8.84 0.83
FL 1828 Validation 11.51 Alive 8.99 8.09 0.72
FL 1829 Validation 4.11 Alive 9.57 8.73 1.08
FL 1830 Validation 5.65 Dead 9.01 8.68 2.25
FL_1833 Training 11.95 Alive 9.74 8.67 0.51
FL 1834 Validation 15.92 Alive 9.22 8.72 1.88
_
FL_1835 Validation , 12.49 Alive 9.26 8.83 2.10
FL_1836 Validation 12.24 Alive 9.55 8.64 0.85
FL 1837 Validation 0.55 Dead 9.47 8.84 1.62
FL_1838 Validation 2.54 Alive 9.90 9.12 1.34
FL_1839 Training 4.48 Alive 8.56 8.32 2.34
FL_1841 Training 0.88 Dead 9.32 9.10 2.66
FL _1842 Validation 4.56 Alive 9.73 8.87 1.07
_
FL_1844 Validation 13.39 Alive 9.41 8.55 0.98
FL_1845 Training 12.92 Dead 9.89 9.04 1.16
FL_1846 Validation 1.80 Dead 9.79 9.61 2.93
FL_1848 Training 12.52 Alive 9.76 8.81 0.82
FL_1851 Training 4.08 Dead 9.43 9.01 2.18
FL_1853 Validation 12.50 Alive 9.28 8.54 1.25
71
CA 2897828 2019-12-20

FL_1854 Validation 13.81 Alive 9.32 8.84 1.98
FL 1855 Validation 9.96 Dead 9.31 8.39 0.75 ,
FL_1857 Validation 8.39 Dead 9.80 9.14 1.65
FL 1861 Validation 3.19 Dead 9.47 8.57 0.88
FL_1862 Validation 7.22 Dead 8.96 8.33 1.44
FL_1863 Validation 10.77 Dead 9.31 8.85 2.00
FL_1864 Training 14.25 Alive 9.98 9.12 1.17
FL_1866 Training 10.72 Dead 9.93 8.94 0.79
FL 1870 , Validation , 6.41 Dead 10.01 9.22 1.36
FL_1873 Training 7.78 Dead 9.39 8.66 1.30
FL_1874 Validation 3.15 Dead 9.38 8.74 1.53
FL 1876 Validation 15.07 Alive 9.59 8.72 , 0.98 ,
FL_1879 Training 7.13 Dead 9.25 8.62 1.53
FL_1880 Validation 12.84 Dead 8.82 8.35 1.82
FL_1882 Training 8.84 Dead 9.43 8.76 , 1.49
FL 1884 Validation 11.92 Dead , 9.48 9.14 2.41
FL_1885 Validation 15.49 Alive 9.70 8.85 1.11
FL_1887 Training 5.14 Dead 9.47 8.57 0.87
FL 1888 Training 15.08 Alive 9.83 8.97 1.11
FL_1890 Training 3.03 Dead 9.29 9.05 2.60
FL_1894 Training 11.37 Dead 9.01 8.64 2.13
FL 1896 Training 12.03 Alive 9.80 8.56 0.08
FL_1897 Training 9.63 Alive 9.02 8.33 1.29
FL_1898 Training 5.20 Alive 8.82 8.25 1.54
FL_1900 Validation 7.38 Alive 9.13 8.26 0.85
FL_1903 Validation , 28.25 , Alive 9.07 8.46 ,
1.54
FL 1904 Validation 7.36 Alive 9.16 8.53 1.50
FL_1905 Validation 3.68 Dead 9.25 8.38 0.87
FL 1906 Training 2.35 Dead 8.04 8.69 4.56
,
FL_1907 Validation 2.35 Dead 8.11 8.21 3.11
FL_1910 Training 13.84 Alive 9.36 8.72 1.56
FL 1912 Validation 0.73 Dead 9.30 9.21 3.02
FL_1913 Training 2.57 Alive 9.77 8.51 0.01 ,
,
FL_1916 Validation 11.61 Alive 9.22 8.49 1.24
FL_1918 Validation 9.95 Dead 9.54 8.77 1.26
FL 1919 Training 10.84 Dead 9.51 8.81 1.44
FL_735 Validation 11.05 Dead 8.81 8.23 1.53
FL_738 Validation 10.15 , Dead 9.19 8.79 2.13
FL 739 Training 10.80 Dead 9.29 8.77 1.85
FL_878 Validation 3.87 Dead 8.85 8.54 2.26
FL_879 Training 4.34 Dead 8.95 8.74 2.56
FL_886 Validation 3.29 Alive 9.43 8.72 1.40
FL_888 Validation 1.32 Dead , 8.76 8.49 , 2.34
FL 1627 Training NA NA 9.60 8.51 0.40
FL 1429 Training NA NA 8.69 8.28 1.93
FL_1850 Validation NA NA 9.75 8.83 , 0.92 ,
FL_1735 Validation NA NA 7.32 8.30 5.24
[00147] In order to visualize the predictive power of the model, the FL
samples were
ranked according to their survival predictor scores and divided into four
quartiles.
Kaplan-Meier plots of overall survival showed clear differences in survival
rate in the
72
CA 2897828 2019-12-20

validation set (Figure 5). The median survival for each of the four quartiles
is set
forth in Table 2362.
Table 2362
Quartile Median survival (years)
1 13.6
2 11.1
3 10.8
4 3.9
[00148] Various clinical variables were found to be significantly associated
with
survival, including the IPI and some of its components and the presence of B-
symptoms. The gene expression-based model was independent of each of these
variables at predicting survival. These clinical variables and the relative
risk of death
associated with each are summarized in Table 2363.
Table 2363
Clinical Criteria % of % of Univariate (clinical
Multivariate (clinical
variable patients' patients' variable only) variable +
survival
relative risk of death predictor score)
among patients in relative risk of death
validation set among patients in
validation set
Training Validation RR2 (95% p-value RR2 (95% p-value
, set set
Age 60 64.5 70.2 1.90 0.044 2.21 (1.48-
<0.001
>60 35.5 29.8 (1.02- 3.29)
3.56)
Stage I-II 33.3 25 1.31 0.447 2.31 (1.51-
<0.001
66.7 75 (0.65- 3.52)
2.64)
Extranodal 2 5.4 20.2 1.58 0.163 2.21 (1.48-
<0.001
sites (#) (0.83- 3.30)
<2 94.6 79.8 2.99)
LDH Normal 77.1 , 66.2 1.77 0.065
2.40 (1.57- <0.001
Greater 22.9 33.8 (0.97- 3.67)
than 3.24)
normal
ECOG 2 9.4 12.5 2.05 0.090 2.17 (1.40-
<0.001
performance (0.89- 3.35)
status <2 90.6 87.5 4.71)
Gender Male 42 65 1.62 0.105 2.17(1.45-
<0.001
Female 58 35 (0.90- 3.25)
2.90)
B-symptoms Present 17.2 21.3 2.05 0.029 2.10 (1.37-
<0.001
(1.08- 3.23)
Absent 82.8 78.7 3.89)
Grade3 1 45 43.4 N/A 0.118 2.55 (1.63-
<0.001
73
CA 2897828 2019-12-20

2 34.8 33.3 2.03 3.99)
(1.04-
3.96)
3 20.2 23.3 1.39
(0.65-
2.98)
Scores 63.1 47.5 N/A 0.029 2.28 (1.46- <0.001
Prognostic 0-1 3.57)
Index4 Scores 33.3 45 2.07
2-3 (1.07-
4.00)
Scores 3.6 7.5 3.73
4-5 (1.18-
11.18)
1 Due to rounding, percentages may not total 100
2 Relative risk of death (RR) based on 2-fold increase in expression
3 RR for grades 2 and 3 calculated with respect to risk of death for grade 1.
The p-
value is calculated for all grades.
4 RR for scores 2-3 and 4-5 calculated with respect to risk of death for
scores 0-1.
The p-value is calculated for all grades.
[00149] The samples in the validation set were divided into three groups based
on
their IPI score, and the relationship between survival and IPI score was
visualized by
Kaplan-Meier plot (Figure 6). Among validation set samples from the low-risk
(IPI 0-
1) and intermediate risk (IPI 2-3) IPI groups, the gene-expression-based
survival
predictor could stratify patients into groups differing by more than 5 years
with
regards to median survival (Figure 7). The high-risk IPI group (IPI 4-5)
comprised
less than 5% of the samples, and was omitted from this analysis. These results
demonstrate that the gene expression-based model is not merely acting as a
surrogate for clinical variables that are known to predict survival in FL, but
rather it
identifies distinct biological attributes of the tumors that are associated
with survival.
Example 4: Development of a second FL survival predictor using gene expression

data from Affvmetrix U133A and U133B microarrays:
[00150] 191 FL were divided into two equivalent groups: a training set (95
samples)
for developing the survival prediction model, and a validation set (96
samples) for
evaluating the reproducibility of the model. Gene expression data from
Affymetrix
U133A and U133B microarrays was obtained for each of the samples. A Cox
74
CA 2897828 2019-12-20

proportional hazards model was used to identify survival predictor genes whose
expression levels were associated with long survival (good prognosis genes) or
short
survival (poor prognosis genes) in the training set. The correlation between
expression and survival for each gene on the microarrays is provided in the
final two
columns of Table 1710. The first of these two columns ("FL_Cox_coefficient")
provides a Cox coefficient indicating the extent to which a 2-fold increase in
expression of a particular gene affects mortality. A positive Cox coefficient
indicates
increasing mortality with increasing expression of the gene, while a negative
Cox
coefficient indicates decreasing mortality with increasing expression of the
gene.
The second of these two columns provides a Cox p-value indicating the
estimated
probability that the increase or decrease in survival associated with the gene
would
occur by chance if there was no connection between the expression of the gene
and
survival.
[00151 ] A hierarchical clustering algorithm (Eisen 1998) was used to identify
gene
expression signatures within the good and poor prognosis genes according to
their
expression pattern across all samples. Eight clusters of coordinately
regulated
genes were observed within the good prognosis gene set and six clusters were
observed in the poor prognosis gene sets. The expression level of every
component
gene in each of these gene expression signatures was averaged to create a gene
expression signature value. After averaging, only ten of the gene expression
signatures were found to be significantly associated with survival in the
training set
(p<0.01). To create a multivariate model of survival, different combinations
of these
ten gene expression signature averages were generated and evaluated for their
ability to predict survival within the training set. Among models consisting
of two
signatures, an exceptionally strong statistical synergy was noted between one
CA 2897828 2019-12-20

signature from the good prognosis group and one from the poor prognosis group.
These gene expression signatures were termed "T-cell" and "macrophage" based
on
the biological function of certain genes within each signature. The T-cell
gene
expression signature included genes that were typically expressed in 1-cells,
while
the macrophage gene expression signature included a number of genes typically
expressed in macrophages. Although these two signatures taken individually
were
not the best predictors of survival, the binary model formed by combining the
two
was more predictive than any combination of three signatures that did not
contain
these two signatures. Using these two signatures as an anchor, other
signatures
were added to the model using a step up procedure (Drapner 1966). Only one of
the remaining eight signatures, termed the B-cell differentiation signature,
contributed significantly to the model in the training set (p=0.054). The B-
cell
differentiation signature included a number of genes that appear to be
involved in B-
cell signal transduction. Table 2364 lists the genes that were used to
generate the
gene expression signature values for the 1-cell, macrophage, and B-cell
differentiation gene expression signatures.
Table 2364
Signature UNIQID Unigene ID Build 167 Gene symbol
B-cell differentiation 1119350 331141 ALDH2
B-cell differentiation 1130922 459987 ANP32B
B-cell differentiation 1130923 459987 ANP32B
B-cell differentiation , 1099291 130774 C9orf105
B-cell differentiation 1102859 446195 FLJ42418
B-cell differentiation 1120976 245644 GCHFR
B-cell differentiation 1098862 303669 M0C26694
B-cell differentiation 1111070 202201
B-cell differentiation 1105935
B-cell differentiation 1139017 274424 NANS
B-cell differentiation 1108988 3532 NLK
B-cell differentiation 1114726 3532 NLK
B-cell differentiation 1097897 266175 PAG
B-cell differentiation 1097901 266175 PAG
B-cell differentiation 1119813 155342 PRKCD
B-cell differentiation 1123298 20191 SIAH2
B-cell differentiation 1101439 63335 TERF2
B-cell differentiation 1120316 63335 TERF2
B-cell differentiation 1096035 105794 UGCGL1
76
CA 2897828 2019-12-20

T-cell 1134945 81897 KIAA1128
T-cell 1134069 405667 CD861
T-cell 1137809 405667 CD8B1
T-cell 1119251 433941 SEPW1
T-cell 1096579 117339 HCST
T-cell 1101004 2969 SKI
T-cell 1137137 195464 FLNA
T-cell 1100871 48353
T-cell 1139461 14770 BIN2
T-cell 1128395 7188 SEMA4C
T-cell 1119880 442844 FMOD
T-cell 1130676 194431 K1AA0992
T-cell 1130668 194431 K1AA0992
T-cell 1135968 119000 ACTN1
T-cell 1097329 528675 TEAD1
T-cell 1098548 436639 NFIC
T-cell 1123038 119000 ACTN1
T-cell 1128356 415792 C1RL
T-cell 1133408 12802 DDEF2
T-cell 1140524 10784 C6orf37
T-cell 1119838 469951 GNAQ
T-cell 1097255 380144
T-cell 1098152 377588 KIAA1450
T-cell 1115194 270737 TNFSF13B
T-cell 1124760 419149 JAM3
T-cell 1120267 256278 TNFRSF1B
T-cell 1137289 36972 CD7
T-cell 1137534 36972 CD7
T-cell 1097307 379754 L0C340061
T-cell 1123613 97087 CD3Z
T-cell 1121720 80642 STAT4
T-cell 1120196 173802 TBC1D4
T-cell 1136087 211576 ITK
1-cell 1132104 173802 TBC1D4
T-cell 1140391 44865 LEF1
T-cell 1098405 362807 IL7R
T-cell 1135743 299558 TNFRSF25
T-cell 1136048 299558 TNFRSF25
T-cell 1123875 428 FLT3LG
T-cell 1098893 43577 ATP8B2
T-cell 1097561 19221 C200rf112
T-cell 1122956 113987 LGALS2
T-cell 1121406 TNFSF12
T-cell 1125532
T-cell 1138538 2014 TRD
T-cell 1103303 49605 C9orf52
T-cell 1119924 32309 INPP1
Macrophage 1123682 114408 TLR5
Macrophage 1099124 355455 SEPT10
Macrophage 1123401 50130 NDN
Macrophage 1134379 150833 C4A
Macrophage 1137481 150833 C4A
Macrophage 1132220 448805 GPRC5B
Macrophage 1119400 181046 DUSP3
Macrophage 1131119 349656 SCARB2
Macrophage 1123566 155935 C3AR1
Macrophage 1138443 77424 FCGR1A
Macrophage 1127943 9641 C1QA
77
CA 2897828 2019-12-20

Macrophage , 1119998 8986 ClQB
Macrophage 1132433 14732 ME1
Macrophage 1119260 18069 LGMN
Macrophage 1098278 166017 MITF
The three signatures were used to generate a survival predictor score using
the
following equation:
Survival predictor score = [2.053*(macrophage gene expression
signature value)] - [2.344*(T-cell gene expression signature value)] -
[0.729*(B-cell differentiation gene expression signature value)].
A higher survival predictor score was associated with worse outcome. According
to
a likelihood ratio test adjusted for the number of variables included, this
model was
significant in predicting survival in both the training set (p=1.8 x 10-8) and
the
validation set (p=2.0 x 10-8). For the 187 FL samples with available clinical
data, the
survival predictor score had a mean of -11.9 and a standard deviation of
0.9418,
with each unit increase in the predictor score corresponding to a 2.5 fold
increase in
the relative risk of death. Data for all 191 samples is shown in Table 2365.
Table 2365
Sample Set B cell T-cell Macrophage Survival
ID # differentiation signature signature predictor
signature value value score
value
FL 1073 Training 9.70 9.14 8.58 -10.89
FL_1074 , Training 11.11 9.06 , 8.52 -11.84
FL_1075 Validation 11.23 8.92 8.75 -11.15
FL 1076 Training 10.02 9.21 8.59 -11.25
FL_1077 Training 9.94 9.77 8.44 -12.82
FL_1078 Training 10.67 9.32 8.21 -12.76
FL 1080 Training 10.62 9.44 8.88 -11.64
FL 1081 Validation 10.38 9.00 8.09 -12.04
FL_1083 Training 10.29 9.77 8.74 -12.47
FL_1085 Validation 9.87 9.24 8.43 -11.55
FL_1086 Validation 10.03 9.50 9.02 -11.06
FL_1087 Training 9.83 9.98 9.37 , -11.31
FL_1088 Validation 10.57 9.21 8.29 -12.27
FL_1089 Training 10.30 9.38 8.27 -12.53
FL 1090 Validation 9.74 9.24 8.20 -11.93
FL_1097 Validation 9.57 , 9.82 8.80 , -11.93
FL 1098 Validation 11.08 9.40 8.97 -11.69
FL_1099 Training 10.23 9.70 9.12 -11.46
78
CA 2897828 2019-12-20

FL_1102 Validation 9.66 9.46 8.90 -10.93
FL 1104 , Training 10.72 9.19 8.20 -12.53
FL 1106 Validation 11.11 9.17 9.57 -9.96
FL_1107 Training 9.70 9.42 9.55 -9.54
FL 1183 Training 9.85 9.25 8.44 -11.54
FL 1184 Training 10.12 9.57 8.86 -11.63
FL 1185 Validation 10.75 9.21 9.13 -10.68
FL 1186 Training 9.76 8.88 8.83 -9.80
FL 1416 Validation 9.94 9.45 8.59 -11.77
FL_1417 Training 10.12 8.53 8.43 -10.08
FL_1418 Validation 9.35 8.86 8.27 -10.59
FL 1419 Training 10.20 9.76 8.53 -12.81
FL 1422 Training 10.22 9.48 8.40 -12.43
FL 1425 Validation 9.61 8.89 8.58 -10.23
FL 1426 Training 10.80 9.06 8.13 -12.41
FL 1427 Training 10.27 8.56 8.13 -10.87
FL 1428 Validation 10.76 9.25 8.38 -12.32
FL_1432 Training 10.51 9.17 9.04 -10.59
FL 1436 Training 9.69 9.40 8.61 -11.42
FL 1440 Training 9.82 9.04 8.21 -11.50
FL 1445 Training 9.24 8.69 8.62 -9.41
FL 1450 Validation 9.70 9.88 10.37 -8.93
FL_1472 Validation 10.78 8.96 8.51 -11.40
FL_1473 Training 9.99 9.70 8.41 -12.75
FL_1474 Validation _ 10.21 9.27 9.05 -10.59
FL_1476 Validation 9.82 9.44 8.78 -11.27
FL 1477 Training , 9.32 9.61 9.03 -10.78
FL_1478 Training 10.19 9.60 8.81 -11.83
FL 1479 Training . 10.69 8.78 9.09 -9.71
FL_1480 Training 10.10 , 9.42 8.70 -11.57
FL_1579 Training 10.15 8.82 8.24 -11.15
FL 1580 Training 10.31 9.59 8.50 -12.54
FL_1581 Training 9.91 8.96 9.05 -9.66
FL_1582 Validation 9.73 8.31 8.06 -10.03
FL 1583 Training 10.95 9.45 8.86 -11.95
FL 1584 Training 9.98 , 9.38 8.46 -11.89
FL_1585 Validation 10.53 8.88 8.46 -11.11
FL_1586 Validation 10.00 9.30 8.42 -11.81
FL 1588 Training 9.59 9.41 8.94 -10.68
FL_1589 Training 10.29 9.68 8.73 -12.27
FL_1591 Training 10.44 9.45 8.56 -12.18
FL 1594 Validation 10.01 9.25 8.56 -11.41
FL_1595 Training 9.61 9.75 9.65 -10.07
FL_1598 Validation 11.18 8.80 8.31 -11.71
FL 1599 Validation 10.55 9.48 8.60 -12.24
FL_1603 Training 9.40 , 9.60 9.77 -9.31
FL_1604 Training 9.92 , 9.21 8.90 -10.54
FL 1606 Validation 9.87 9.45 9.17 , -10.52
FL 1607 Validation 9.76 9.37 8.50 -11.63
FL_1608 Validation 9.92 _ 8.90 8.39 -10.85
FL 1610 Validation 10.02 9.38 9.74 -9.30
FL_1611 Validation 10.18 9.41 8.69 -11.64
FL_1616 Training 9.62 9.33 8.85 -10.71
FL_1617 Validation 9.90 _ 8.95 8.39 -10.98
FL 1619 Validation 9.98 9.37 8.47 -11.85
FL_1620 Validation 9.43 8.95 8.12 -11.19
FL_1622 Training 9.84 _ 9.15 8.31 -11.56
FL_1623 Training 9.95 9.61 8.97 -11.37
79
CA 2897828 2019-12-20

FL_1624 Validation 10.55 9.06 8.43 -11.61
FL_1625 Validation 10.00 8.89 8.23 -11.22 s
FL 1626 Validation 11.05 8.62 8.10 -11.62
FL_1628 Validation 10.08 9.81 8.66 -12.57
FL_1637 Validation 9.77 9.95 9.59 -10.76
FL 1638 Validation 10.25 9.20 9.07 -10.41
FL_1639 Training 10.29 9.52 8.99 -11.35
FL_1643 Training 9.80 9.72 9.00 -11.46
FL 1644 Validation 9.51 9.46 8.61 -11.43
FL 1645 Training 9.39 9.46 8.70 -11.15
FL_1646 Training 9.90 9.25 8.52 -11.42
FL 1647 Training 9.51 9.12 8.95 -9.92
FL 1648 Training 10.02 9.18 7.86 -12.67
FL-_1652 Training 9.62 9.39 9.19 -10.16
FL 1654 Validation 10.32 8.59 8.10 -11.02
FL 1655 Training 10.12 9.53 8.75 -11.74
FL_1656 Validation 10.54 9.08 8.55 -11.42
FL_1657 Training 10.53 9.53 8.55 -12.46
FL 1660 Training 10.24 8.75 8.27 -10.99
FL_1661 Validation 10.08 9.85 9.00 -11.97
FL_1662 Validation 9.85 9.56 9.49 -10.11
FL_1664 Validation 10.16 9.35 8.48 -11.92
FL 1669 Training 9.48 8.76 8.28 -10.45
FL_1670 Training 9.76 9.66 9.66 -9.92
FL_1675 Training 10.57 9.28 8.41 -12.18
FL 1681 Validation 10.48 9.52 8.66 -12.19
FL_1683 Validation , 9.88 9.92 9.07 -11.83
FL_1684 Training 9.64 9.53 8.85 -11.20
FL 1716 Validation 9.90 8.91 8.22 -11.23
FL_1717 Validation 9.87 9.34 8.95 -10.71
FL_1718 Training 10.00 9.21 7.98 -12.49
FL 1719 Validation 9.87 9.06 8.42 -11.14
FL_1720 Training 10.70 8.77 8.92 -10.05
FL_1729 Training 10.50 9.23 8.65 -11.53
FL_1732 Validation 9.91 7.68 8.54 -7.69
FL 1761 Validation 9.81 9.22 8.39 -11.54
FL_1764 Training 9.81 9.24 8.77 -10.80
FL_1768 Training 10.12 9.36 8.50 -11.86
FL_1771 Training 9.92 9.12 8.68 -10.79
FL_1772 , Validation _ 9.72 9.42 8.43 -11.87
FL 1788 Training 9.65 9.05 9.12 -9.51
FL_1790 Training 9.58 9.83 9.48 -10.56
FL 1792 Validation , 9.79 9.29 8.67 -11.11
FL_1795 Training 9.58 10.18 9.33 -11.69
FL 1797 Validation 9.93 9.26 8.79 -10.90
FL_1799 Training 10.49 9.28 8.64 -11.65
FL_1810 Validation 10.06 8.55 8.21 -10.52
FL 1811 Validation 9.84 9.37 8.08 -12.56
FL-_1825 Training 10.49 9.44 9.03 -11.24
FL_1827 Training 10.06 9.76 8.84 -12.08
FL_1828 Validation 10.55 8.93 7.67 -12.87
FL 1829 Validation 9.85 9.58 8.65 -11.87
FL_1830 Validation 10.80 8.99 8.67 -11.15
FL_1833 Training 10.41 9.83 8.82 -12.52
FL_1834 Validation 10.81 9.25 8.63 -11.85
FL 1835 Validation 9.36 - 9.25 8.91 -10.21
FL_1836 Validation 10.58 9.58 8.61 -12.50
FL 1837 Validation 10.22 9.47 8.76 -11.68
CA 2897828 2019-12-20

FL_1838 Validation 10.51 9.89 9.19 , -11.98
FL 1839 Training 10.79 8.54 8.19 -11.09
FL_1841 Training 10.32 9.31 9.18 -10.48
FL_1842 Validation 10.36 9.69 8.92 -11.95
FL 1844 Validation 10.92 9.43 . 8.49 -12.65
FL_1845 Training 9.87 9.87 9.06 -11.73
FL_1846 Validation 9.66 9.81 9.93 -9.63
FL_1848 Training 9.82 9.74 8.70 -12.14
FL_1851 Training 9.89 9.47 9.03 -10.87
FL_1853 Validation 9.96 9.28 8.54 -11.49
FL 1854 Validation 9.97 9.29 8.73 -11.12
FL 1855 Validation 9.95 9.33 8.42 -11.85
FL_1857 Validation 10.35 9.81 9.28 -11.50
FL_1861 Validation 9.73 9.46 8.43 -11.96
FL 1862 Validation 10.42 8.94 8.22 -11.69
FL 1863 Validation 10.79 9.29 8.82 -11.54
FL:1864 Training 9.67 9.97 9.07 -11.80
FL 1866 Training 10.19 9.88 8.89 -12.33
FL 1870 Validation 9.78 10.07 9.30 -11.63
FL:1873 Training 10.09 9.41 8.77 -11.40
FL 1874 Validation 10.05 9.33 8.69 -11.37
FL:1876 Validation 10.15 9.59 8.67 -12.08
FL_1879 Training 9.73 9.21 8.58 -11.06
FL_1880 Validation 10.02 8.79 8.35 -10.77
FL 1882 Training 9.59 9.44 8.80 -11.05
FL_1884 Validation 9.76 9.51 9.26 -10.38
FL_1885 Validation 10.48 9.66 8.75 -12.32
FL 1887 Training 9.98 9.42 8.47 -11.96
FL 1888 Training 9.73 9.83 8.99 -11.67
FL_1890 Training 10.06 9.33 8.98 -10.76
FL_1894 Training 9.85 8.99 8.75 -10.29
FL_1896 Training 10.21 9.80 8.51 -12.94
FL_1897 Training 10.67 8.99 8.26 -11.90
FL_1898 Training 9.59 8.77 8.21 -10.68
FL 1900 Validation 10.12 9.10 8.10 -12.08
FL_1903 Validation 11.08 8.99 8.39 -11.93
FL_1904 Validation 10.20 9.16 8.30 -11.87
FL 1905 Validation 9.73 9.21 8.22 -11.80
FL_1906 Training 9.95 8.15 8.44 -9.01
FL_1907 Validation 10.12 7.95 7.99 -9.62
FL_1910 Training 11.03 9.38 8.74 -12.10
FL_1912 Validation 9.83 9.38 9.36 -9.95
FL_1913 Training 9.81 9.75 8.43 -12.69
FL_1916 Validation 9.83 9.18 8.40 -11.43
FL_1918 Validation 9.86 9.52 8.79 -11.45
FL 1919 Training 9.87 9.53 8.79 -11.48
FL 735 Validation 10.48 8.73 8.23 -11.20
FL:738 Validation 11.05 9.10 8.75 -11.43
FL_739 Training 9.66 9.25 8.74 -10.78
FL_878 Validation 10.61 8.92 8.65 -10.89
FL_879 Training 9.92 8.94 8.78 -10.14
FL_886 Validation 10.16 9.41 8.63 -11.73
FL_888 Validation 9.35 8.76 8.38 -10.15
FL_1627 Training 9.82 9.48 8.49 -11.94
FL 1429 Training 10.06 8.70 8.14 -11.01
FL_1850 Validation 9.58 9.73 8.70 -11.93
FL_1735 Validation 9.60 7.46 8.42 -7.19
81
CA 2897828 2019-12-20

[0015211n order to visualize the predictive power of the model, the FL samples
were
ranked according to their survival predictor scores and divided into four
quartiles.
Kaplan-Meier plots of overall survival showed clear differences in survival
rate in the
validation set (Figure 8). The median survival for each of the four quartiles
is set
forth in Table 2366.
Table 2366
Quartile Median survival (yrs.) 5-year survival 10-
year survival
1 NR 94% 79%
2 11.6 82% 62%
3 8.8 69% 39%
4 3.9 38% 22%
Example 5: Development of a third FL survival predictor using gene expression
data
from the Lymph Dx microarrav:
[00153] 191 FL samples were divided into two equivalent groups: a training set
for
developing the survival prediction model, and a validation set for evaluating
the
reproducibility of the model. Gene expression data from the Lymph Dx
microarray
was obtained for those genes listed in Table 2364, above. This gene expression
data was used to calculate gene expression signature values for the
macrophage, T-
cell, and B-cell differentiation gene expression signatures, and these
signature
values were used to generate a survival predictor score using the following
equation:
Survival predictor score = [1.51*(macrophage gene expression
signature value)] ¨ [2.11*(T-cell gene expression signature value)] ¨
[0.505*(13-cell differentiation gene expression signature value)].
A higher survival predictor score was associated with worse outcome. For the
187
FL samples with available clinical data, the survival predictor score had a
mean of
10.1 and a standard deviation of 0.69, with each unit increase in the
predictor score
corresponding to a 2.7 fold increase in the relative risk of death. Data for
all 191
samples is shown in Table 2367.
82
CA 2897828 2019-12-20

Table 2367
Sample Set B cell T-cell Macrophage Survival
ID # differentiation signature signature
predictor
signature value value score
value
FL_1073 Training 8.26 8.17 7.36 -10.30
FL 1074 Training 9.53 8.12 7.56 -10.53
FL_1075 Validation 9.81 8.00 7.99 -9.77
FL_1076 Training 8.46 8.10 7.62 -9.86
FL_1077 Training 8.45 8.66 7.32 -11.49
FL_1078 , Training 9.23 8.32 7.32 -11.18
FL_1080 Training 9.18 8.37 7.86 -10.42
FL_1081 Validation 8.96 8.01 6.94 -10.96
FL 1083 Training 8.72 8.65 7.89 -10.75
FL_1085 Validation 8.34 8.17 7.54 -10.07
FL_1086 Validation 8.50 8.35 7.94 -9.94
FL_1087 Training 8.02 8.88 8.48 -10.00
FL_1088 Validation 9.10 8.15 7.38 -10.65
FL_1089 Training 8.76 8.31 7.35 -10.86
FL_1090 Validation 8.18 8.23 7.43 -10.28
FL 1097 Validation 8.07 8.81 7.90 -10.73
FL_1098 Validation 9.53 8.30 8.09 -10.11
FL_1099 Training 8.44 8.56 8.26 -9.86
FL 1102 Validation 7.92 8.43 7.94 -9.80
FL_1104 Training 9.17 8.07 7.21 -10.78
FL_1106 Validation 9.71 8.15 8.77 -8.85
FL_1107 Training 8.16 8.44 8.60 -8.95
FL_1183 Training 8.49 8.15 7.23 -10.56
FL_1184 Training 8.81 8.49 7.91 -10.43
FL_1185 Validation 9.31 8.19 8.06 -9.80
FL 1186 Training 8.43 7.87 7.83 -9.04 ,
FL_1416 Validation 8.42 8.34 7.63 -10.34
FL_1417 Training 8.65 7.51 7.05 -9.58
FL 1418 Validation 7.96 7.82 7.22 -9.62
FL_1419 Training 8.80 8.71 7.55 -11.43
FL_1422 Training 8.63 8.35 7.39 -10.83
FL_1425 Validation 8.21 7.92 7.62 -9.36
FL 1426 Training 9.39 8.09 7.15 -11.01
FL_1427 Training 8.66 7.51 7.00 -9.65
FL_1428 Validation 9.33 8.18 7.39 -10.81
FL 1432 Training 8.98 8.17 7.93 -9.81
FL_1436 Training 8.04 8.17 7.35 -10.20
FL 1440 Training 8.29 7.82 7.15 -9.89
FL 1445 Training 8.04 7.78 7.63 -8.94
FL_1450 Validation 8.25 8.81 9.52 -8.39
FL_1472 Validation 9.29 7.88 7.33 -10.26
FL 1473 Training 8.49 8.57 7.52 -11.03
FL_1474 Validation 8.59 8.09 8.53 -8.54
FL_1476 Validation 8.25 8.39 7.71 -10.23
FL 1477 Training 7.94 8.57 7.88 -10.21
FL_1478 Training 8.57 8.40 7.88 -10.16
FL_1479 Training 9.15 7.83 7.87 -9.27
FL_1480 Training 8.25 8.38 7.44 -10.63
FL 1579 Training 8.70 7.73 7.43 -9.48
FL_1580 Training 8.86 8.46 7.64 , -10.79
FL_1581 Training 8.41 7.89 8.08 -8.69
FL_1582 Validation 8.20 7.42 6.99 -9.24
FL_1583 Training 9.34 8.34 7.94 -10.32
. 83
CA 2897828 2019-12-20

FL_1584 Training 8.50 8.33 7.75 -10.17
FL 1585 Validation 9.08 7.96 7.72 -9.72
FL 1586 Validation 8.52 8.25 7.36 -10.61
FL_1588 Training 7.97 8.35 7.73 -9.98
FL_1589 Training 8.85 8.48 7.76 -10.66
FL 1591 Training 8.92 8.36 7.77 -10.42
FL11594 Validation 8.54 8.22 7.74 -9.96
FL 1595 Training 8.05 8.82 8.68 -9.57
FL_1598 Validation 9.74 7.81 6.97 -10.88
FL_1599 Validation 9.13 8.42 7.69 -10.77
FL_1603 Training 7.97 8.66 8.90 -8.86
FL_1604 Training 8.47 8.14 7.75 -9.75
_ FL 1606 Validation 8.34 8.32 8.11 -9.51
FL_1607 Validation 8.33 8.30 7.39 -10.57
FL_1608 Validation 8.35 7.88 6.98 -10.31
FL_1610 Validation 8.48 8.35 8.86 -8.52
FL_1611 Validation 8.54 8.33 7.64 -10.37
FL_1616 Training 8.03 8.39 7.67 -10.18
FL_1617 Validation 8.30 7.85 7.52 -9.40
FL 1619 Validation 8.53 8.31 7.64 -10.32
FL_1620 Validation 8.09 7.99 7.17 -10.11
FL_1622 Training 8.14 8.10 7.36 -10.09
FL 1623 Training 8.45 8.52 8.15 -9.93
FL_1624 Validation 9.13 8.12 7.46 -10.49
FL_1625 Validation 8.53 7.94 7.17 -10.23
FL 1626 Validation 9.63 7.67 7.17 -10.22
FL_1628 Validation 8.63 8.76 7.95 -10.86
FL_1637 Validation 8.07 8.81 8.79 -9.38
FL 1638 Validation 8.52 8.18 8.19 -9.18
FL 1639 Training 8.70 8.33 7.89 -10.06
FL 1643 Training 8.26 8.62 8.01 -10.26
FL_1644 Validation 8.28 8.33 7.77 -10.02
FL 1645 Training 7.84 8.32 7.68 -9.91
FL_1646 Training 8.40 8.26 7.71 -10.01
FL_1647 Training 8.10 8.04 7.92 -9.10
FL_1648 Training 8.33 8.08 6.87 -10.90
FL_1652 Training 8.15 8.33 8.37 -9.07
FL 1654 Validation 8.67 7.62 7.03 -9.85
FL_1655 Training 8.53 8.41 7.75 -10.36
FL 1656 Validation 9.09 8.09 7.62 -10.16
FL_1657 Training 8.95 8.44 7.58 -10.89
FL_1660 Training 8.82 7.79 7.26 -9.93
FL 1661 Validation 8.56 8.79 8.17 -10.53
FL_1662 Validation 8.30 8.47 8.69 -8.93
FL_1664 Validation 8.62 8.23 7.56 -10.31
FL 1669 Training 7.89 7.67 7.39 -9.02
FL_1670 Training 8.01 8.54 8.64 -9.03
FL_1675 Training 9.00 8.21 7.36 -10.76
_ FL 1681 Validation 8.83 8.39 7.59 -10.72
FL_1683 Validation 8.14 8.85 7.97 -10.74
FL_1684 Training 7.99 8.42 7.84 -9.97
FL_1716 Validation 8.28 7.90 7.26 -9.88
FL 1717 Validation 8.27 8.21 7.89 -9.60
FL_1718 Training 8.50 8.17 7.15 -10.75
FL_1719 Validation 8.35 8.02 7.21 -10.26
FL_1720 Training 9.03 7.65 8.01 -8.61
FL_1729 Training 8.97 8.27 7.69 -10.37
FL_1732 Validation 8.49 6.82 7.71 -7.02
84
CA 2897828 2019-12-20

FL_1761 Validation 8.36 8.19 7.29 -10.49
FL 1764 Training 8.52 8.24 7.94 -9.69
FL_1768 Training 8.70 8.25 7.63 -10.28
FL_1771 Training 8.55 8.19 7.65 -10.04
FL 1772 Validation 8.30 8.38 7.41 -10.71
FL_1788 Training 8.14 8.06 8.11 -8.87
FL 1790 Training 7.95 8.69 8.36 -9.74
FL_1792 Validation 8.16 8.20 7.64 -9.88
FL 1795 Training 7.94 9.08 8.37 -10.54
FL 1797 Validation 8.17 8.21 7.87 -9.57
FL 1799 Training 9.02 8.21 7.77 -10.14
FL 1810 Validation , 8.43 7.52 7.06 -9.47
FL 1811 Validation 8.33 8.24 , 7.07 -10.93
FL11825 Training 8.90 8.39 7.97 -10.18
FL 1827 Training 8.47 8.77 7.96 -10.76
FL_1828 Validation 9.13 7.87 6.76 -11.01
FL 1829 Validation 8.34 8.51 7.59 -10.71
FL:1830 Validation 9.26 , 8.04 7.62 -10.13
FL 1833 Training 8.82 8.86 7.88 -11.26
FL_1834 Validation 9.25 8.17 , 7.62 -10.39
FL 1835 Validation 7.71 8.16 8.01 -9.02
FL 1836 Validation 9.06 8.52 7.59 -11.09
FL_1837 Validation 8.57 8.33 7.37 -10.79
FL 1838 Validation 8.78 8.72 8.04 -10.69
FL_1839 Training 9.27 7.36 7.37 -9.08
FL 1841 Training 8.66 8.35 8.17 -9.64
FL 1842 Validation , 8.62 8.50 8.02 -10.19
FL_1844 Validation 9.37 8.40 7.47 -11.18
FL 1845 Training 8.33 8.84 8.30 -10.32
FL_1846 Validation 8.11 8.75 9.06 -8.89
FL_1848 Training 8.19 8.60 7.91 -10.33
= FL 1851 Training 8.37 8.50 8.15 -9.84
FL_1853 Validation 8.37 8.14 7.43 -10.19
FL_1854 Validation _ 8.50 8.29 7.96 -9.78
FL 1855 Validation 8.63 8.34 7.54 -10.58
FL_1857 Validation 8.73 8.82 8.45 -10.26
FL_1861 , Validation 8.21 8.50 7.50 -10.77
FL_1862 Validation 8.98 , 7.96 7.31 -10,28
FL 1863 Validation 9.30 8.22 7.86 -10.18
FL 1864 Training 8.13 8.93 8.27 -10.46
FL 1866 Training _ 8.62 8.78 7.91 -10.93
FL 1870 Validation 8.16 8.97 8.52 -10.18
FL_1873 Training 8.55 8.30 8.00 -9.74
FL_1874 Validation 8.43 , 8.20 7.59 -10.10
FL 1876 Validation 8.48 8.52 7.70 -10.64
FL_1879 Training 8.29 8.21 7.66 -9.94
FL 1880 Validation 8.56 7.76 7.34 -9.61
FL 1882 Training 8.02 8.40 7.71 -10.14
FL 1884 Validation 8.14 8.46 8.42 -9.24
FL 1885 Validation , 8.88 8.57 7.78 -10.81
FL 1887 Training 8.38 8.39 7.38 -10.78
FL_1888 Training 8.14 - 8.74 8.07 -10.37
FL_1890 Training 8.45 8.24 8.11 -9.41
FL_1894 Training 8,38 7.97 7.82 -9.25
FL 1896 Training 8.63 8.71 7.52 -11.37
FL_1897 Training 9.01 7.91 6.93 -10.78
FL_1898 Training 8.08 7.75 7.09 -9.74
FL_1900 Validation 8.61 7.94 6.84 -10.77
CA 2897828 2019-12-20

FL_1903 Validation 9.63 7.96 7.30 -10.64
FL 1904 Validation 8.79 8.14 7.15 -10.82
FL_1905 Validation 8.22 8.24 7.36 -10.43
FL_1906 Training 8.40 7.40 7.24 -8.93
FL_1907 Validation 8.61 7.11 6.59 -9.40
FL_1910 , Training 9.47 8.28 7.63 -10.73
FL_1912 Validation 8.32 8.45 8.52 -9.18
FL_1913 Training 8.24 8.60 7.23 -11.41
FL 1916 Validation 8.31 8.04 7.27 -10.19
FL_1918 Validation 8.30 8.49 7.78 -10.37
FL 1919 Training 8.05 8.42 8.00 -9.75
FL _735 Validation 9.03 7.83 7.41 -9.88
FL_738 Validation 9.54 8.07 7.65 -10.30
FL_739 Training 8.14 8.09 7.69 -9.57
FL_878 Validation 9.17 7.91 7.70 -9.69
FL 879 Training 8.37 7.96 7.67 -9.45
FL_886 Validation 8.59 8.38 7.67 -10.44
FL 888 Validation 7.85 7.71 7.07 -9.56
FL -1627 Training 8.26 8.17 7.36 -10.30
FL_1429 Training 9.53 8.12 7.56 -10.53
FL_1850 Validation 9.81 8.00 7.99 -9.77
FL_1735 Validation 8.46 8.10 7.62 -9.86
[00154] In order to visualize the predictive power of the model, the FL
samples were
ranked according to their survival predictor scores and divided into four
quartiles.
Kaplan-Meier plots of overall survival showed clear differences in survival
rate in the
validation set (Figure 9).
Example 6: Development of a first DLBCL survival predictor using gene
expression
data from Affvmetrix U133A and U133B microarrays:
(00155] Gene expression data from Affymetrix U133A and U133B microarrays was
obtained for 231 DLBCL samples. The follow-up time and status at follow-up for
each of the subjects from whom these samples were acquired is listed in Table
2368. Table 2368 also indicates which samples were used in creating the
survival
predictor.
Table 2368
Sample ID # Length of follow- Status at
follow-up Used in creating survival
up (years) predictor?
ABC_1000 0.69 Dead Yes
ABC 1002 0.28 Dead Yes
ABC_1023 5.57 Dead Yes
ABC_1027 0.25 Dead Yes
ABC_1031 6.64 Dead Yes
86
CA 2897828 2019-12-20

ABC 1034 2.31 Dead Yes
ABC_1038 0.71 Dead Yes
ABC 1043 2.31 Dead Yes
ABC 1045 2.26 Dead Yes
ABC 1055 7.81 Alive Yes
ABC_1057 2.13 Dead Yes
ABC_1059 2.00 Dead Yes
ABC_1061 1.04 Dead Yes
ABC_1946 0.68 Dead No
ABC_1994 1.21 Dead No
ABC 2001 1.32 Dead No
ABC 304 1.31 Dead Yes
ABC_305 0.82 Alive Yes
ABC_309 2.80 Alive Yes
ABC 413 0.60 Dead Yes
ABC:428 11.38 Alive Yes
ABC_432 0.38 Dead Yes
ABC 446 2.82 Dead Yes
ABC 462 7.49 Dead Yes
ABC 477 1.70 Dead Yes
ABC:481 10.75 Alive Yes
ABC_482 7.72 Alive Yes
ABC 538 0.34 Dead Yes
ABC_541 4.11 Alive Yes
ABC_544 1.31 Dead Yes
ABC_547 0.05 Dead Yes
ABC 577 1.65 Alive Yes
ABC:616 0.99 Dead Yes
ABC 626 2.49 Dead Yes
ABC_633 2.02 Alive Yes
ABC_642 0.34 Dead Yes
ABC 644 0.31 Dead Yes
ABC 645 6.08 Dead Yes
ABC:646 2.59 Dead Yes
ABC 651 2.34 Alive Yes
ABC:652 0.01 Dead Yes
ABC_660 0.20 Dead Yes
ABC 663 0.62 Dead Yes
ABC:668 6.44 Alive Yes
ABC 676 1.00 Dead Yes
ABC 678 0.06 Dead Yes
ABC 687 0.94 Dead Yes
ABC 689 2.54 Dead Yes
ABC 692 10.53 Alive Yes
ABC 694 4.83 Alive Yes
ABC_700 5.40 Dead Yes
ABC_702 4.13 Dead Yes
ABC 704 9.67 Alive Yes
ABC_709 0.47 Dead Yes
ABC 712 3.26 Dead Yes
ABC 714 2.45 Dead Yes
ABC_717 0.42 Dead Yes
ABC_725 0.96 Dead Yes
ABC_726 7.62 Alive Yes
ABC 730 1.03 Dead Yes
ABC_753 0.04 Dead Yes
ABC 756 7.21 Alive Yes
ABC_771 6.80 Dead Yes
87
CA 2897828 2019-12-20

ABC_779 0.35 Dead Yes
ABC 800 0.33 Dead Yes
ABC_807 0.31 Dead Yes
ABC_809 0.51 Dead Yes
ABC_816 1.86 Dead Yes
ABC_820 1.59 Dead Yes
ABC_823 0.16 Dead Yes
ABC_835 1.22 Dead Yes
ABC_839 0.29 Dead Yes
ABC_841 10.14 Alive Yes
ABC_858 3.58 Dead Yes
ABC_872 5,00 Alive Yes
ABC 875 8.45 Alive Yes
ABC:912 16.79 Alive Yes
ABC 996 0.21 Dead Yes
GCB_1005 5.77 Alive Yes
GCB_1008 6.46 Alive Yes
GCB 1009 9.68 Alive Yes
GCB_1021 14.59 Alive Yes
GCB_1025 2.86 Dead Yes
GCB 1026 6.94 Dead Yes
GCB_1037 0.23 Dead Yes
GCB_1039 2.05 Dead Yes
GC B_1049 1.33 Dead Yes
GCB_1051 0.12 . Dead Yes
GCB_1058 0.42 Dead Yes
,
GCB_1060 6.45 Alive Yes
GCB_1990 0.06 Dead No
,
GCB_1991 1.01 Dead No
GCB_2017 0.08 Dead No
GCB 2018 0.17 Dead No
_
GCB 2095 0.97 Alive No
GCB-_412 12.12 Alive Yes
GCB 415 5.38 . Dead Yes
GCB_421 1.24 Dead Yes
GCB_424 10.62 Dead Yes
GCB 433 0.76 Dead Yes
GCB_434 10.53 Alive Yes
GCB_438 8.15 Alive Yes
GCB_459 9.65 Alive Yes
GCB 470 11.17 _ Alive Yes
GCB_479 7.24 Alive Yes
GCB_492 11.29 Alive Yes
,
GCB_517 , 3.03 Dead Yes
GCB_523 8.36 Alive Yes
GCB_524 5.88 Alive Yes
GCB_529 1.06 Dead Yes
GCB_533 0.71 Dead Yes
GCB_537 4.99 Dead Yes
GCB_543 3.47 _ Alive Yes
GCB_545 1.10 Dead Yes
GCB 549 2.68 Dead Yes
GCB 550 21.78 Alive Yes
GCB_553 0.82 Dead Yes
GCB_565 9.11 Dead Yes .
GCB_572 14.24 Alive Yes
,
GCB 617 5.88 Alive Yes
GCB_618 5.65 Alive Yes
88
CA 2897828 2019-12-20

GCB_619 8.76 Alive Yes
GCB 623 2.43 Alive Yes
GCB 627 1.27 Dead Yes
GCB_654 7.37 Alive Yes
GCB 661 0.56 Alive Yes
GCB 669 7.11 Alive Yes
GCB 672 6.78 Alive Yes
GCB 674 7.22 Alive Yes
GCB 675 6.02 Alive Yes
GCB 681 9.70 Alive Yes
GCB_688 0.33 Dead Yes
GCB 695 0.15 Dead Yes
GCB_698 3.88 Alive Yes
GCB 701 3.90 Alive Yes
GCB 710 1.08 Dead Yes
GCB_711 3.93 Dead Yes
GCB_722 3.32 Alive Yes
GCB 724 1.40 Dead Yes
GCB 731 10.18 Alive Yes
GCB_742 4.09 Alive Yes
GCB 744 8.86 Alive Yes
GCB_745 1.33 Dead Yes
GCB 747 15.41 Alive Yes
GCB_749 10.40 Alive Yes
GCB 758 1.10 Dead Yes
GCB_772 2.48 Alive Yes
GCB_777 4.27 Dead Yes
GCB 792 . 5.53 Alive Yes
GCB 795 3.43 Alive Yes
GCB_797 6.87 Dead Yes
GCB 803 1.45 Dead Yes
GCB 810 11.72 Alive Yes .
GCB_817 2.76 Dead Yes
GCB 818 0.10 Dead Yes
GCB_819 0.72 Dead Yes
GCB_821 9.47 Alive Yes
GCB_832 4.01 Alive Yes
GCB 836 4.29 Alive Yes
GCB 840 3.40 Alive Yes
GC B_847 4.16 Alive Yes
GCB 860 3.03 Dead Yes
GCB_871 0.41 Dead Yes
GCB_874 0.12 Dead Yes
GCB 995 6.65 Alive Yes
PMBL_1006 7.12 Alive Yes
PMBL_1024 19.83 Alive Yes
PMBL 1048 7.70 Alive Yes
PMBL_1053 1.04 Dead Yes
PMBL_1920 1.97 Alive No
PMBL 1921 4.16 Alive No
PMBL_1923 1.60 Alive No
PMBL_1924 6.11 Alive No
PMBL_1935 12.42 Alive No
PMBL 1941 0.71 Alive No
PMBL_1942 0.88 Alive No
PMBL_1943 8.96 Alive No
PMBL_1945 0.84 Dead No
PMBL_1948 7.96 Alive No
89
CA 2897828 2019-12-20

PMBL 1949 4.28 Alive No
PMBL 1989 1.33 Dead No
PMBL 1992 1.00 Dead No
PMBL 1993 1.33 Dead No
PMBL 2002 6.62 Alive No
PMBL 2019 0.99 Dead No
PMBL 2020 2.08 Alive No
PMBL 2092 1.27 Alive No
PMBL 484 1.40 Dead . Yes
PMBL 546 0.78 Dead Yes
PMBL_570 14.40 Alive Yes
PMBL 621 8.14 Alive Yes
PMBL 638 0.70 Dead Yes
PMBL 691 0.32 Dead Yes
PMBL 791 1.33 Dead Yes
PMBL 824 12.24 Alive Yes
PMBL_906 16.80 Alive Yes
PMBL 994 4.79 Alive Yes
PMBL 998 9.11 Alive Yes
UC DLBCL_1001 0.33 Dead Yes
UC_DLBCL 1004 6.72 Alive Yes
UC_DLBCL:1007 2.26 Dead Yes
UC_DLBCL 1018 0.03 Dead Yes
UC_DLBCL_1041 3.13 Dead Yes .
UC DLBCL 1054 12.34 Alive Yes
UC_DLBCL _306 2.69 Alive Yes
UC_DLBCL 310 0.97 Alive Yes
UC_DLBC1_1449 9.16 Alive . Yes
UC DLBCL_452 9.17 Alive Yes
UC_DLBCL 458 1.18 Dead Yes
UC_DLBCL_460 9.02 Alive Yes
UC_DLBCL_491 4.47 Dead Yes
UC DLBCL 528 , 1.64 Alive Yes
UC_DLBCL_615 4.94 Alive Yes .
UC_DLBCL_625 5.24 Alive . Yes
UC_DLBCL_664 0.62 Dead Yes
UC_DLBCL_671 3.35 Alive Yes
UC_DLBCL_682 0.11 Dead Yes
UC_DLBCL 683 , 7.42 Alive Yes
UC DLBCL:684 1.92 Dead Yes
UC_DLBCL_748 1.01 Dead Yes
UC_DLBCL_751 9.99 Alive Yes
UC_DLBCL_808 0.37 Dead Yes
UC_DLBCL_831 11.02 Dead Yes
UC_DLBCL 834 1.64 Dead Yes
.
UC_DLBCL:838 0.00 Dead Yes
UC_DLBCL_851 0.05 Dead Yes .
UC_DLBCL_854 1.51 Dead Yes
UC DLBCL 855 1.67 Alive Yes
UC_DLBCL_856 0.60 Dead Yes
[00156] The correlation between expression of each gene represented on the
microarrays and survival was estimated using a Cox proportional hazards model.
The results of this survival analysis are provided in the final two columns of
Table
CA 2897828 2019-12-20

1723. The first of these two columns ("DLBCL_Cox_coefficient") provides a Cox
coefficient indicating the extent to which a 2-fold increase in expression of
a
particular gene affects mortality. A positive Cox coefficient indicates
increasing
mortality with increasing expression of the gene, while a negative Cox
coefficient
indicates decreasing mortality with increasing expression of the gene. The
second
of these two columns ("DLBCL_Cox_P_value") provides a Cox p-value indicating
the
estimated probability that the increase or decrease in survival associated
with the
gene would occur by chance if there was no connection between the expression
of
the gene and survival.
[00157] Genes that were significantly correlated with survival (p<0.001) were
grouped into gene expression signatures using a hierarchical clustering
algorithm.
The expression level of every component gene in each of these gene expression
signatures was averaged for each sample to create a gene expression signature
value. A step-up procedure (Drapner 1966) was applied to determine the optimal
number of gene signatures to use in the survival predictor model. First, the
gene
expression signature that was most significantly associated with survival was
included in the model. Next, the gene expression signature with the second
highest
association with survival was added to the model to form a two-component
model.
This procedure was repeated until there was no gene expression signature to
add to
the model with a p-value of <0.05.
[00158] The final prediction model incorporated gene expression signature
values
from three gene expression signatures. The first gene expression signature
added
to the model was termed "ABC DLBCL high," because it included genes that were
more highly expressed in ABC than in GCB (Rosenwald 2002). The second gene
expression signature added to the model was termed "lymph node," because it
91
CA 2897828 2019-12-20

reflected the response of non-tumor cells in the lymph node to the malignant
lymphoma cells. The final gene expression signature added to the model was
termed "MHC class II," because it included all of the genes encoding the MHC
class
II alpha and beta chains. Table 2369 shows the genes that were averaged to
form
each of these signatures.
Table 2369
Signature UNIQID Gene symbol Survival p-value
ABC DLBCL high 1134271 POU5F1 3.09E-05
ABC DLBCL high 1121564 DRIL1 4.06E-05
ABC DLBCL high 1119889 PDCD4 7.28E-05
ABC DLBCL high 1133300 CTH 1.23E-04
ABC DLBCL high 1106030 MGC:50789 1.70E-04
ABC DLBCL high 1139301 FLJ20150 4.49E-04
ABC DLBCL high 1122131 CHST7 5.18E-04
ABC DLBCL high 1114824 LIMD1 5.20E-04
ABC DLBCL high 1100161 _ L0C142678 6.24E-04
ABC DLBCL high 1120129 TLE1 6.95E-04
Lymph node 1097126 TEM8 5.14E-09
Lymph node 1120880 LTBP2 9.80E-07
Lymph node 1098898 FLJ31066 1.09E-06
_ Lymph node 1123376 RARRES2 1.68E-06
,
Lymph node 1128945 SLC12A8 2.90E-06
Lymph node 1130994 DPYSL3 3.37E-06
_ Lymph node 1124429 SULF1 3.53E-06
Lymph node 1099358 FLJ39971 4.09E-06
Lymph node 1130509 SPARC 6.23E-06
_ Lymph node 1095985 TMEPAI 7.07E-06
Lymph node 1123038 ACTN1 7.90E-06
. Lymph node 1133700 CDH11 8.20E-06
_ Lymph node 1122101 TFEC 9.66E-06
_
Lymph node 1124296 SDC2 9.99E-06
MHC Class II 1123127 HLA-DRA 1.21E-06
MHC Class II 1136777 HLA-DQA1 3.45E-06
_
MHC Class II 1137771 HLA-DRB1 3.95E-06
MHC Class ll 1134281 HLA-DRB4 2.70E-05
MHC Class II 1136573 HLA-DPA1 2.92E-05
MHC Class II 1132710 HLA-DRB3 7.09E-05
[00159] Fitting the Cox proportional hazards model to the three gene
expression
signature values resulted in the following model:
Survival predictor score = [0.586*(ABC DLBCL high gene expression
signature value)] - [0.468*(lymph node gene expression signature
value)] - [0.336*(MHC Class II gene expression signature value)].
92
CA 2897828 2019-12-20

A higher survival predictor score was associated with worse outcome. According
to
a likelihood ratio test adjusted for the number of variables included, this
model was
significant in predicting survival at p=2.13x10-13. In order to visualize the
predictive
power of the model, the 205 samples used to create the model were ranked
according to their survival predictor scores and divided into four quartiles.
Kaplan-
Meier plots of overall survival probability show clear differences in survival
rate
between these four quartiles (Figure 10). The five-year survival probabilities
for
each quartile are set forth in Table 2370.
Table 2370
Quartile 5-year survival
1 83%
2 59%
3 33%
4 17%
93
CA 2897828 2019-12-20

Example 7: Development of a second DLBCL survival predictor using gene
expression data from the Lymph Dx microarraT
[00160]A DLBCL survival model based on gene expression had been developed
previously using proliferation, germinal center B-cell, lymph node, and MHC
class II
gene expression signatures and the expression of the single gene BMP-6
(Rosenwald 2002). BMP-6 expression was poorly measured on the Lymph Dx
microarray, but genes associated with each of these four gene expression
signatures exhibited associations with survival similar to those observed
using
Lymphochip microarrays. DLBCL samples were divided into two groups: a training
set (100 samples) for developing the survival prediction model, and a
validation set
(100 samples) for evaluating the reproducibility of the model. Gene expressed
in the
training set samples were clustered, and lymph node, germinal center B-cell,
MHC
class II, and proliferation gene expression signatures were identified. Within
each
signature, expression of genes that were associated with survival (p<0.01) was
averaged to generate a gene expression signature value for each signature.
Table
2371 lists the genes that were used to generate the gene expression signature
value
for each signature.
Table 2371
Signature UNIQID Unigene ID Build 167 Gene symbol
Germinal center B-cell 1099686 117721
Germinal center B-cell 1099711 243596
Germinal center B-cell 1103390 271752 BPNT1
Germinal center B-cell 1106025 49500 KIAA0746
Germinal center B-cell 1128287 300063 ASB13
Germinal center B-cell 1132520 283063 LMO2
Germinal center B-cell 1138192 126608 NR3C1
Germinal center B-cell 1529318 291954
Germinal center B-cell 1529344 317970 SERPINA11
Germinal center B-cell 1529352 446195
Germinal center B-cell 1096570 409813 ANUBL1
Germinal center B-cell 1097897 266175 PAG
Germinal center B-cell 1097901 266175 PAG
Germinal center B-cell 1098611 433611 PDK1
Germinal center B-cell 1100581 155024 BCL6
Germinal center B-cell 1115034 387222 NEK6
94
CA 2897828 2019-12-20

Germinal center B-cell 1120090 155024 BCL6
Germinal center B-cell 1120946 25209 MAPK10
Germinal center B-cell 1121248 54089 BARD1
Germinal center B-cell 1123105 434281 PTK2
Germinal center B-cell 1125456 300592 MYBL1
Germinal center B-cell 1128694 171466 ELL3
Germinal center B-cell 1128787 114611 C7orf10
Germinal center B-cell 1132122 307734 MME
Germinal center B-cell 1136269 101474 MAST2
Germinal center B-cell 1136702 155584 KIAA0121
Germinal center B-cell 1139230 29724 PLEKHF2
Germinal center B-cell 1529292 NA
Germinal center B-cell 1529295 116441
Lymph node 1097126 274520 ANTXR1
Lymph node 1099028 334838 FNDC1
Lymph node 1099358 93135
Lymph node 1101478 146246 MGC45780
Lymph node 1103497 50115
Lymph node 1121029 412999 CSTA
Lymph node 1124429 409602 SULF1
Lymph node 1135068 71719 PDLIM3
Lymph node 1136051 520937 CSF2RA
Lymph node 1136172 38084 SULT1C1
MHC class II 1136777 387679 HLA-DQA1
MHC class II 1136877 409934 HLA-DQB1
Proliferation 1096903 437460 FLJ10385
Proliferation 1120583 153768 RNU3IP2
Proliferation 1123289 5409 POLR1C
Proliferation 1131808 75447 RALBP1
Proliferation 1133102 360041 FRDA
Proliferation 1136595 404814 VDAC1
[00161] Table 2372 lists p-values for the association of each signature with
survival
in the training set, the validation set, and overall.
Table 2372
Signature Training set Validation set Overall
Lymph node 4.0 x 10-5 2.3 x 10-8 6.8 x 1045
Proliferation 8.1 x 10-5 3.4 x 10-3 2.1 x 10-8
Germinal center B-cell 6.2 x 10-8 2.1 x 10-3 5.0 x 10-8
MHC class II 2.4 x 10-2 2.7 x 10-3 3.1 x 10-4
[00162] The four gene expression signatures were used to generate a survival
predictor score using the following equation:
Survival predictor score = [-0.4337*(lymph node gene expression
signature value)] + [0.09*(proliferation gene expression signature
CA 2897828 2019-12-20

value)] - [0.4144*(germinal center B-cell gene expression signature
value)] - [0.2006*(MHC class II gene expression signature value)].
A higher survival predictor score was associated with worse outcome. For the
200
DLBCL samples used to generate the model, the survival predictor score had a
mean of 5.7 and a standard deviation of 0.78, with each unit increase in the
predictor score corresponding to an approximately 2.7 fold increase in the
relative
risk of death. Data for all 200 samples is presented in Table 2373.
Table 2373
Sample ID # Set Lymph Proliferation Germinal
MHC Survival
node signature center B- class II
predictor
signature value cell signature score
value signature value
value
ABC 1000 Validation 6.50 8.92 7.60 11.50 -5.08
ABC_1002 Validation 7.00 _ 8.58 7.27
12.54 -5.50
ABC _1023 Validation 7.43 8.99 6.80 11.42 -5.05
ABC_1027 Training 5.68 9.00 6.87 12.31 -4.70
ABC_1031 Validation 8.02 9.00 7.17 11.68 -5.53
ABC _1034 Validation 6.06 9.61 6.72 11.83 -4.58
ABC_1038 Training 6.83 8.97 7.17 12.30 -5.23
ABC 1043 Training 6.96 9.01 6.77 12.29 -5.11
ABC_1045 Validation 8.18 8.21 6.77 12.07 -5.66
ABC_1055 Validation 5.58 9.16 7.30 13.05 -4.76
ABC 1057 Training 7.33 8.94 7.74 12.05 -5.53
ABC 1059 Validation 9.02 8.46 7.15 11.35 -6.08
_
ABC 1061 Training 7.13 9.18 7.09 12.28 -5.21
ABC- 304 Validation 5.92 8.80 6.76 12.76 -4.84
ABC_305 Training 5.92 , 8.74 7.50 11.89 -4.91
ABC 309 Validation 8.86 8.39 7.62 12.53 -6.46
ABC-_413 Validation 6.45 9.32 6.55 9.04 -4.16
ABC 428 Training 7.52 9.19 _ 7.98 10.25 -5.51
ABC 432 Validation 6.48 9.33 7.45 9.56 -4.56
ABC 446 Training 7.91 9.42 7.41 10.55 -5.46
A BC:462 Validation 6.41 8.85 6.67 13.36 -5.03
ABC 477 Validation 6.26 9.02 6.69 12.45 -4.89
ABC_481 Training 8.18 8.30 7.35 11.98 -5.91
ABC 482 Training 8.59 9.01 7.66 12.35 -6.16
ABC 538 Validation 8.06 8.84 _ 7.17 11.83 -5.69
ABC 541 Training 6.14 8.52 7.42 10.59 -4.71
ABC 544 Training 6.91 9.03 6.82 11.87 -4.89
A BC_547 Validation 5.80 8.96 7.14 11.38 -4.60
ABC_577 Validation 7.84 8.65 8.16 11.95 -5.94
ABC_616 Validation , 6.03 9.05 , 7.36 12.64 -4.84
ABC 626 , Validation 7.48 9.22 7.25 11.11 -5.27
ABC_633 Training 7.74 8.35 7.39 12.45 -5.80
ABC_642 Training 5.71 8.82 6.41 13.80 -4.62
ABC 644 Validation 6.64 9.15 7.05 13.28 -5.20
A BC_645 Training 8.44 8.81 7.93 13.39 -6.43
ABC_646 Validation 5.94 9.11 6.71 11.60 -4.63
96
CA 2897828 2019-12-20

ABC_652 Validation 5.87 8.85 6.88 12.73 -4.77
ABC 660 Training 5.19 9.34 6.64 10.17 -3.86
ABC 663 Training 5.69 9.02 7.33 12.82 -4.91
ABC_668 Validation 7.12 9.28 7.03 10.57 -4.91
ABC_676 Training 4.95 8.90 7.09 13.32 -4.61
ABC 678 Training 5.84 9.11 7.34 11.26 -4.41
ABC_687 Validation 5.15 9.89 6.56 10.46 -3.76
ABC_689 Training 6.49 8.86 7.10 12.56 -4.88
ABC 692 Validation 7.32 8.96 7.25 11.57 -5.32
ABC_694 Validation 8.28 9.21 8.01 12.41 -6.23
ABC_700 Training 7.29 8.97 7.55 12.10 -5.48
ABC_702 Validation 7.60 8.66 6.86 12.55 -5.45
ABC_704 Training 7.07 8.92 7.03 12.83 -5.35
ABC_709 Validation 5.92 8.58 6.37 13.40 -4.66
ABC 712 Validation 5.79 9.12 6.34 , 12.02 -
4.23
ABC 714 Training , 7.49 8.88 7.49 11.97 -5.54
ABC 717 Training 7.17 9.45 7.01 11.34 -5.05
ABC_725 Training 6.71 9.01 6.52 12.76 -4.86
ABC 726 Validation 6.91 8.72 6.71 11.91 -4.90
ABC 730 Validation 6.28 9.22 7.28 12.14 -4.88
ABC_753 Training 6.84 9.64 7.05 13.00 -5.22
ABC_756 Training 7.67 8.45 7.59 12.48 -5.85
ABC 771 Training 6.98 8.76 6.91 12.20 -5.18
ABC_779 Training 6.73 9.32 6.78 9.82 -4.44
ABC_800 Validation 8.75 8.31 7.45 11.91 -6.04
ABC 807 Training 5.50 9.53 6.92 7.56 -3.79
ABC_809 Training 7.40 8.70 7.68 10.83 -5.50
ABC_816 Training 5.20 9.91 7.65 10.64 -4.14
ABC 820 Training 6.71 8.94 6.55 11.98 -4.85
ABC_823 Validation 5.58 9.26 6.44 10.09 -3.97
ABC_835 Validation 6.95 8.68 8.04 12.31 -5.59
ABC 839 Training 6.63 9.17 7.23 11.89 -5.04
ABC_841 Validation 6.35 9.51 7.52 13.19 -5.28
ABC_858 Training 7.63 8.51 7.12 11.74 -5.42
ABC_872 Training 6.78 8.73 7.41 12.47 -5.44
ABC 875 Training 7.59 8.81 7.20 11.26 -5.25
ABC_912 Validation 7.01 8.55 7.45 12.79 -5.64
A BC_996 Validation 5.00 9.53 6.70 10.02 -3.94
GCB_1005 Validation 8.28 8.67 9.11 13.27 -6.98
GCB_1008 Training 8.17 8.59 9.83 12.83 -7.06
GCB 1009 Training 6.63 9.02 10.07 12.28 -6.19
GCB_1021 Validation 6.44 8.83 9.34 13.20 -6.15
GCB_1025 _ Validation 7.87 8.48 9.27 12.37 -6.57
GCB_1026 Training 7.71 8.30 9.81 13.52 -6.85
GCB 1037 Training 4.95 8.83 9.35 12.57 -5.22
GCB_1039 Training 7.63 8.65 9.01 13.28 -6.47
GCB_1049 Validation 8.54 8.61 8.12 12.60 -6.41
GCB_1051 Validation 6.26 9.09 9.48 12.76 -5.97
GCB 1058 Validation 7.12 8.89 8.34 12.80 -5,85
GCB_1060 Validation 8.27 8.84 8.94 12.96 -6.75
GCB_412 Training 7.22 8.33 8.50 13.09 -6.09
GCB 415 Training 9.01 8.62 8.38 11.99 -6.47
GCB_421 Training 7.59 7.89 7.49 12.20 -5.80
GC 6_424 Training 9.29 8.42 8.51 12.44 -6.79
GCB_433 Training 8.45 8.34 8.02 12.64 -6.54
GCB 434 Training 8.46 8.55 9.17 12.54 -6.98
GCB_438 Validation 8.14 8.71 9.13 12.51 -6.67
GCB_459 Validation 8.98 8.39 8.42 11.37 -6.49
97
CA 2897828 2019-12-20

GCB_470 Validation 7.72 8.57 8.67 12.23 -6.12
GCB 479 Validation 6.86 8.25 7.13 13.07 -5.35
GCB_492 Training 8.01 8.61 9.51 12.34 -6.63
GCB_517 Validation 8.57 8.73 7.99 12.76 -6.48
GCB 523 Training 5.96 8.56 8.74 12.77 -5.72
GCB_524 Training 8.51 8.09 8.76 12.51 __ -6.57
GCB_529 Training 5.12 9.17 8.88 10.77 __ -4.86
GCB_533 Training 8.88 8.81 8.36 12.44 -6.60
GCB 537 Validation 7.42 8.19 9.73 13.29 -6.68
GCB 543 Validation 8.49 8.02 8.66 12.06 -6.45
GCB_545 Training 8.65 8.28 6.90 12.90 -6.13
GCB 549 Validation 6.87 8.24 8.65 12.15 -6.00
GCB_550 Validation 8.98 8.29 8.76 12.24 -6.94
GCB_553 Validation 8.51 8.64 8.62 12.63 -6.69
GCB_565 Validation 7.97 8.79 9.79 13.42 -6.98
GCB 572 Training 7.61 8.60 9.39 12.58 -6.42
GCB_617 Validation 8.31 7.89 7.54 13.17 -6.12
GCB_618 Training 5.66 8.97 9.20 13.32 -5.54
GCB_619 Validation 7.83 8.65 9.34 12.12 -6.36
GCB 623 Training 7.16 8.88 9.26 12.35 -6.21
GCB_627 Validation 8.13 8.83 8.62 11.85 -6.31
GCB 654 Training 6.30 9.60 8.45 10.00 -4.88
GCB_661 Validation 8.46 8.51 8.18 12.66 -6.33
GCB_669 Training 7.88 8.65 8.59 12.32 -6.19
GCB 672 Training 8.29 8.61 8.14 12.41 -6.21
GOB 674 Validation 8.36 8.62 7.76 12.33 -6.14
GCB_675 Validation 6.01 9.52 8.90 10.12 -5.09
GCB_681 Training 9.25 8.72 , 8.72 12.59
-6.89
GCB 688 Validation 6.97 9.01 9.90 9.94 -5.99
GCB_695 Validation 8.80 8.73 9.23 12.45 -6.84
GCB_698 Validation 9.27 8.35 8.85 11.99 ,
-6.96
GCB 701 Training 7.77 7.93 8.68 13.10 -6.33
GCB_710 Validation 6.12 8.78 7.65 13.19 -5.24
GCB 711 Training 7.57 8.80 8.43 11.44 -5.84
GCB 722 Training 7.78 8.31 , 8.93 12.61
-6.51
GCB_724 Training 7.88 9.08 8.74 11.53 -6.21
GCB_731 Validation 7.72 8.92 9.08 12.20 -6.46
GCB_742 Validation 8.33 8.55 8.58 12.95 -6.70
GCB_744 Training 8.02 8.64 9.36 11.85 -6.52
GCB_745 Training 8.47 8.34 8.93 11.95 -6.67
GCB_747 Validation 7.64 8.48 8.32 13.06 -6.27
GCB_749 Training 7.57 8.61 9.40 12.55 -6.56
GCB_758 Validation 5.66 8.77 7.89 12.51 -4.63
GCB_772 Validation 8.52 7.81 7.95 12.25 -6.34
GCB_777 Validation 7.52 8.65 8.57 11.69 -6.10
GCB_792 Training 8.14 8.64 9.21 12.08 -6.65
GCB_795 Validation 9.19 8.17 8.81 11.60 -6.92
GCB_797 Validation 7.50 8.62 8.08 12.84 -6.09
GCB_803 Validation 6.19 8.65 9.49 13.18 -6.11
GCB_810 Training 8.46 8.32 8.10 13.13 -6.50
GCB 817 Training 6.93 8.51 9.49 11.09 -6.04
GCB_818 Training 7.18 8.96 , 8.08 12.23
-5.76
GCB_819 Validation 7.16 8.97 8.06 13.22 -5.79
G0B_821 Validation 8.13 8.59 8.90 12.41 -6.61
GCB 832 Training 7.83 8.35 8.71 12.47 -6.37
GCB_836 Validation 7.84 8.99 8.50 11.46 -5.85
GCB_840 Training 8.24 7.75 7.40 11.74 -5.77
GCB_847 Training 7.82 8.17 8.97 12.55 -6.51
98
CA 2897828 2019-12-20

GCB 860 Training 7.12 8.39 9.34 11.54 -6.10
GCB 871 Training 5.59 9.60 7.28 11.16 -4.23
GCB 874 Training 8.53 9.14 8.95 11.65 -6.47
GC 6_995 Validation 6.98 8.68 8.54 12.22 -5.76
PMBL_1006 Validation 7.34 8.51 7.66 10.94 -5.33
PMBL 1024 Validation 7.62 8.48 8.56 10.89 -5.96
PMBL_1048 Validation 8.68 8.16 7.23 12.18 -6.08
PMBL 1053 Training 7.02 8.28 8.24 11.12 -5.31
PMBL _484 Training 7.15 8.45 7.01 13.62 -5.41
PMBL 546 Validation 8.19 7.88 7.66 11.73 -6.06
PMBL-_570 Training 9.34 8.21 8.48 12.70 -6.86
PMBL_621 Training 8.08 8.60 9.14 12.96 -6.72
PMBL 638 Training 7.56 8.26 8.00 11.37 -5.75
PMBL_691 Validation 6.48 8.92 8.40 10.17 -5.04
PMBL_ 791 Validation 7.72 8.65 8.94 11.56 -6.16
PMBL 824 Validation , 8.06 8.01 7.76 13.28 -6.11
PMBL_994 Training _ 9.15 8.36 7.46 12.43 -6.29
PM BL 998 Training 6.70 8.35 9.24 13.19 -6.20
UC DLBCL 1001 Validation 6.74 8.43 7.10 12.76 -5.31
UC_DLBCL_1004 Validation 7.54 8.75 8.01 13.09 -6.10
UC DLBCL_1007 Training 9.97 8.44 7.64 12.97 -6.85
UC_DLBCL_1018 Training 6.42 8.38 6.97 12.71 -5.03
UC DLBCL 1041 Validation 5.76 8.69 6.78 13.38 -4.71
UC DLBCL 1054 Training 8.92 8.65 8.51 11.48 -6.59
UC-_DLBCC 306 Validation 7.85 8.90 8.31 12.36 -6.23
UC DLBCL 310 Training 8.14 8.80 7.63 12.27 -6.03
UC_DLBCL_449 Validation 9.03 8.48 7.07 12.17 -6.01
UC_DLBCL_458 Training 5.92 8.53 8.28 9.60 -4.96
UC_DLBCL 460 Validation 7.92 9.08 8.30 12.29 -6.13
UC_DLBCL_491 Training 7.65 8.33 7.35 12.39 -5.53
UC_DLBCL_528 Validation 6.99 8.56 7.36 11.63 -5.35
UC_DLBCL_615 Validation 7.11 8.32 8.77 12.80 -6.10
UC DLBCL 625 Training 8.93 7.78 7.85 12.62 -6.46
UC_DLBCL_664 Training 7.62 8.15 8.17 12.72 -6.04
UC_DLBCL_671 Training 8.09 8.48 7.61 11.53 -5.78
UC DLBCL 682 Training 7.38 8.35 7.14 12.33 -5.43
UC_DLBCL_683 Training 7.91 8.36 7.78 12.57 -6.02
UC_DLBCL_684 Validation 8.06 8.63 8.29 12.76 -6.29
UC_DL8CL_748 Validation 5.38 8.57 7.45 9.55 -4.23
UC_DLBCL_751 Training 6.33 8.65 8.88 13.14 , -5.74
UC_DLBCL 808 Training 7.42 9.01 7.44 13.09 -5.63
UC_DLBCL-_831 Validation 8.33 8.30 7.46 11.58 -5.84
UC_DLBCL_834 Training 6.98 9.09 8.61 11.77 -5.66
UC_DLBCL_838 Validation 7.25 8.40 7.23 12.56 -5.36
UC_DLBCL_851 Validation 6.28 9.05 6.78 8.19 -4.10
UC_DLBCL_854 Validation 7.36 8.50 7.39 12.59 -5.53
UC_DLBCL_855 Training 8.31 7.94 7.49 12.08 -6.07
UC_DLBCL_856 Validation 5.65 9.01 8.52 9.32 -4.68
[00163] In order to visualize the predictive power of the model, the 200
samples
were ranked according to their survival predictor scores and divided into four
quartiles. Kaplan-Meier plots of overall survival probability show clear
differences in
survival rate between these four quartiles (Figure 11).
99
CA 2897828 2019-12-20

Example 8: Development of a third DLBCL survival predictor using gene
expression
data from the Lymph Dx microarrav:
[00164] The number of genes used to generate the DLBCL survival predictor in
Example 7 was reduced in order to create a survival predictor compatible with
RT-
PCR. The list of genes from the lymph node and germinal center B-cell gene
expression signatures was narrowed to those three genes from each signature
that
were most closely correlated with the lymph node and germinal center B-cell
gene
expression signature values, respectively. The genes from the proliferation
gene
expression signature did not add significantly to the reduced gene survival
prediction
model, so they were removed entirely. The expression of the genes within each
signature was averaged on the 10g2 scale to generate a gene expression
signature
value for each signature. Table 2374 lists the genes that were used to
generate
these gene expression signature values.
Table 2374
Signature UNIQID Unigene ID Build 167 Gene symbol
Germinal center B-cell 1099686 117721
Germinal center B-cell 1529318 291954
Germinal center B-cell 1529344 317970 SERPINA11
Lymph node 1097126 274520 ANTXR1
Lymph node 1099358 93135
Lymph node 1121029 412999 CSTA
MHC class II 1136777 387679 HLA-DQA1
MHC class II 1136877 409934 HLA-DQB1
[00165] Table 2375 lists p-values for the association of each signature with
survival
in the training set, the validation set, and overall.
Table 2375
Signature Training set Validation set Overall
Lymph node 6.1 x 10-6 0.0021 2.1 x 10-17
Germinal center B-cell 3.5 x 10-4 0.0099 2.7 x 10-5
MHC class II 0.024 0.0026 0.00031
[00166] The three gene expression signatures were used to generate a survival
predictor score using the following equation:
100
CA 2897828 2019-12-20

Survival predictor score = [-0.32*(lymph node gene expression
signature value)] - [0.176*(germinal center B-cell gene expression
signature value)] - [0.206*(MHC class II gene expression signature
value)].
A higher survival predictor score was associated with worse outcome. For the
200
DLBCL samples used to generate the model, the survival predictor score had a
mean of 6.54 and a standard deviation of 0.69, with each unit increase in the
predictor score corresponding to an approximately 2.7 fold increase in the
relative
risk of death. Data for all 200 samples is presented in Table 2376.
Table 2376
Sample ID # Set Lymph Germinal MHC class Survival
node center B-cell II predictor
signature signature signature score
value value value
ABC 1000 Validation 8.08 5.68 11.50 -5.96
ABC 1002 Validation 8.32 6.06 12.54 -6.31
ABC_1023 Validation 9.36 4.74 11.42 -6.18
ABC_1027 Training 7.41 4.90 12.31 -5.77
ABC_1031 Validation 9.40 5.23 11.68 -6.33
_ ABC 1034 Validation 7.47 4.92 11.83 -5.69
_
ABC 1038 Training 7.89 5.84 12.30 -6.09
ABC_1043 Training 7.84 4.66 12.29 -5.86
ABC_1045 Validation 9.31 4.66 12.07 -6.29
ABC 1055 Validation _ 6.46 6.38 13.05 -5.88
ABC 1057 Training 9.13 7.93 12.05 -6.80
ABC_1059 Validation 10.93 4.82 11.35 -6.68
ABC 1061 Training 8.18 5.04 12.28 -6.04
ABC- 304 Validation 7.31 6.47 12.76 -6.10
ABC 305 Training 7.02 6.60 11.89 -5.86
ABC 309 Validation 10.47 7.00 12.53 -7.16
ABC 413 Validation 7.99 4.80 9.04 -5.26
ABC_428 Training 9.43 7.59 10.25 -6.47
ABC_432 Validation 7.29 8.16 9.56 -5.74
ABC 446 Training 9.49 5.46 10.55 -6.17
ABC_462 Validation 7.72 4.97 13.36 -6.10 ,
ABC 477 Validation 7.16 3.69 12.45 -5.51
ABC 481 Training 9.75 6.89 11.98 -6.80
ABC_482 Training , 10.51 , 7.64 12.35 -7.25
ABC_538 Validation 8.79 5.00 11.83 -6.13
ABC_541 Training 7.70 5.80 10.59 -5.67
ABC 544 Training 8.90 3.98 11.87 -5.99
ABC_547 Validation 7.05 5.18 11.38 -5.51
ABC 577 Validation 9.93 8.05 11.95 -7.06
ABC 616 Validation 7.34 4.54 12.64 -5.75
ABC 626 Validation 8.78 6.77 11.11 -6.29
ABC_633 Training 9.63 5.02 12.45 -6.53
101
CA 2897828 2019-12-20

ABC_642 Training 7.31 4.95 13.80 -6.05
ABC 644 Validation 7.72 5.35 13.28 -6.15
ABC 645 Training 9.77 6.21 13.39 -6.98
ABC_646 Validation 7.39 3.75 11.60 -5.41
ABC 652 Validation 7.51 4.53 12.73 -5.82
ABC 660 Training 5.85 3.55 10.17 -4.59
ABC:663 Training 7.04 5.06 12.82 -5.78
ABC_668 Validation 8.00 5.65 10.57 -5.73
ABC 676 Training 6.53 4.29 13.32 -5.59
ABC_678 Training 6.87 7.48 11.26 -5.83
ABC_687 Validation 6.39 3.78 10.46 -4.87
ABC 689 Training 8.29 5.07 12.56 -6.13
ABC:692 Validation 8.10 5.26 11.57 -5.90
ABC_694 Validation 9.67 8.15 12.41 -7.09
ABC_700 Training , 8.37 6.75 12.10 -6.36
ABC 702 Validation 8.44 4.59 12.55 -6.09
ABC_704 Training 8.51 4.34 12.83 -6.13
ABC 709 Validation 7.47 4.54 13.40 -5.95
ABC 712 Validation 7.12 3.99 12.02 -5.46
ABC 714 Training 9.57 7.03 11.97 -6.77
ABC_717 Training 8.33 5.54 11.34 -5.98
ABC_725 Training 8.04 4.40 12.76 -5.97
ABC 726 Validation 7.79 4.18 11.91 -5.68
ABC_730 Validation 8.13 7.36 12.14 -6.40
ABC_753 Training 9.24 6.60 13.00 -6.80
ABC 756 Training 9.51 5.21 12.48 -6.53
ABC_771 Training 8.08 4.74 12.20 -5.93
ABC_779 , Training 8.11 4.09 9.82 -5.34
ABC 800 Validation 10.34 4.83 11.91 -6.61
ABC_807 Training 6.58 4.44 7.56 -4.44
ABC_809 Training 9.29 5.72 10.83 -6.21
ABC_816 Training 6.36 6.36 10.64 -5.35
ABC 820 Training 8.10 4.79 11.98 -5.90
ABC_823 Validation 6.63 4.85 10.09 -5.05
ABC_835 Validation 9.17 7.78 12.31 -6.84
ABC 839 Training 8.06 4.97 11.89 -5.90
ABC_841 Validation 8.05 , 6.24 13.19 -
6.39
ABC_858 Training 9.02 4.86 _ 11.74 -6.16
ABC 872 Training 8.67 5.85 12.47 -6.37
ABC_875 Training 9.60 5.59 11.26 -6.37
ABC 912 Validation 7.99 7.74 12.79 -6.56
ABC 996 Validation 6.89 6.23 " 10.02 -5.36
GCB_ 1005 Validation 9.02 9.56 13.27 -7.30
GCB_1008 Training 9.27 10.49 12.83 -7.46
GCB 1009 Training 7.80 10.09 12.28 -6.80
GCB_1021 Validation , 8.73 9.20 13.20 -7.13
GCB_1025 Validation , 9.94 9.97 12.37 -7.49
GCB_1026 Training 9.54 10.20 13.52 -7.63
GCB 1037 Training 6.34 8.79 12.57 -6.17
GCB_1039 Training 8.71 9.94 , 13.28 -7.27
GCB 1049 Validation 10.53 8.18 12.60 -7.41
GCB 1051 Validation 7.63 10.18 12.76 -6.86
GCB_1058 Validation 8.61 9.04 12.80 -6.98
GCB 1060 Validation 10.23 9.38 12.96 -7.59
GCB1412 Training 8.79 7.92 13.09 -6.90
GCB 415 Training 10.72 8.57 11.99 -7.41
GCB_421 Training 9.23 5.26 12.20 -6.39
GCB_424 Training 11.14 8.46 12.44 -7.62
102
CA 2897828 2019-12-20

GCB 433 Training 9.26 8.52 12.64 -7.07
GCB 434 Training 9.73 10.13 12.54 -7.48
GCB_438 Validation 9.60 9.99 12.51 -7.41
GCB_459 Validation 10.51 7.75 11.37 -7.07
GCB 470 Validation 9.56 6.63 12.23 -6.74
_
GCB_479 Validation 7.77 4.71 13.07 -6.01
GCB_492 Training 8.82 9.52 12.34 -7.04
GCB_517 Validation 9.92 6.96 12.76 -7.03
GCB_523 Training 6.59 9.17 12.77 -6.35
GCB_524 Training 10.00 7.83 12.51 -7.16
GC8_529 Training 5.61 7.93 10.77 -5.41
GCB 533 Training 9.55 5.54 , 12.44 -6.59
GCB 537 Validation 8.25 10.25 13.29 -7.18
GCB:543 Validation 9.92 8.85 12.06 -7.21
GCB 545 Training 9.69 4.91 12.90 -6.62
GCB_549 Validation 7.86 8.88 12.15 -6.58
GCB_550 Validation 10.64 9.53 12.24 -7.60
GCB_553 Validation 10.14 9.05 12.63 -7.44
GCB 565 Validation 9.08 10.80 13.42 -7.57
GCB 572 Training 8.93 10.03 12.58 -7.21
GCB:617 Validation 9.27 7.80 13.17 -7.05
GCB 618 Training 7.23 9.11 13.32 -6.66
GCB_619 Validation 9.63 9.63 12.12 -7.27
GC B_623 Training 8.94 9.07 12.35 -7.00
GCB 627 Validation 9.72 8.33 11.85 -7.02
GCB 654 Training 7.04 5.60 10.00 -5.30
GCB:661 Validation 10.27 7.92 12.66 -7.29
GOB 669 Training 9.15 9.29 12.32 -7.10
GCB 672 Training 9.69 7.36 12.41 -6.95
GCB_674 Validation 9.93 6.23 12.33 -6.81
GCB_675 Validation 7.48 8.46 10.12 -5.97
GCB 681 Training 10.77 9.52 12.59 -7.72
GCB_688 Validation 8.01 10.17 9.94 -6.40
GCB_695 Validation 10.58 9.38 12.45 -7.60
GC B_698 Validation 10.44 9.00 11.99 -7.39
GCB_701 Training 9.38 9.27 13.10 -7.33
GCB_710 Validation 6.96 5.59 13.19 -5.93
GCB_711 Training 9.28 8.49 11.44 -6.82
GCB_722 Training 8.93 9.51 12.61 -7.13
GCB_724 Training 9.51 8.39 11.53 -6.90
GCB_731 Validation 8.82 9.19 12.20 -6.95
GCB_742 Validation 9.95 9.37 12.95 -7.50
GCB_744 Training 10.23 10.11 11.85 -7.49
GCB_745 Training 10.29 9.71 11.95 -7.46
GCB_747 Validation 9.83 9.79 13.06 -7.56
GCB_749 Training 8.57 10.27 12.55 -7.14
GCB_758 Validation 6.88 5.69 12.51 -5.78
GCB_772 Validation 9.92 7.28 , .. 12.25 .. -6.98
GCB_777 Validation 9.03 9.63 11.69 -6.99
GCB_792 Training 9.49 9.06 12.08 -7.12
GCB 795 Validation 11.12 9.02 11.60 -7.54
GCB_797 Validation 8.42 5.90 12.84 -6.38
GCB 803 Validation 7.33 10.11 , .. 13.18 .. -6.84
GCB_810 Training 10.00 8.22 13.13 -7.35
GCB 817 Training 8.60 10.16 11.09 -6.82
GCB_818 Training 9.14 7.78 12.23 -6.81
GCB_819 Validation 9.08 8.63 , 13.22 -7.15
GOB 821 Validation 10.05 9.81 12.41 -7.50
103
CA 2897828 2019-12-20

GCB_832 Training 8.83 6.91 12.47 -6.61
GCB_836 Validation 9.49 7.86 11.46 -6.78
GCB_840 Training 9.45 5.02 11.74 -6.33
GCB_847 Training 9.41 8.77 12.55 -7.14
GCB_860 Training 9.02 6.66 11.54 -6.43
GCB_871 Training 6.60 4.46 11.16 -5.20
GCB_874 Training 10.39 9.13 11.65 -7.33
GCB_995 Validation 8.52 9.35 12.22 -6.89
PMBL 1006 Validation 8.72 4.67 10.94 -5.86
PM BL_1024 Validation 9.30 8.47 10.89 -6.71
PM BL_1048 Validation 10.30 4.98 12.18 -6.68
PM BL 1053 Training 8.75 9.78 11.12 -6.81
PMBL 484 Training 8.25 4.96 13.62 -6.32
PMBL_546 Validation 9.66 6.07 11.73 -6.57
PMBL_570 Training 10.58 8.54 12.70 -7.50
PMBL 621 Training 9.39 9.94 12.96 -7.43
PMBL_638 Training 9.81 8.35 11.37 -6.95
PMBL_691 Validation 8.37 7.51 10.17 -6.10
PMBL 791 Validation 9.29 8.65 11.56 -6.88
PMBL_ 824 Validation 9.87 7.19 13.28 -7.16
PMBL_994 Training 11.27 6.73 12.43 -7.35
PMBL_998 Training 7.92 8.34 13.19 -6.72
UC _ DLBCL_ 1001 Validation 8.25 5.63 12.76 -6.26
UC _ DLBCL _1004 Validation 9.01 7.01 13.09 -6.81
UC_DLBCL_1007 Training 11.42 6.73 12.97 -7.51
UC_DLBCL_1018 Training 7.77 4.58 12.71 -5.91
UC _DLBCL 1041 Validation 7.90 4.33 13.38 -6.05
UC DLBCL 1054 Training 10.41 . , 8.72 11.48 -7.23
UC-_ DLBCE 306 Validation 9.42 6.54 12.36 -6.71
UC DLBCL 310 Training 9.97 5.50 12.27 -6.69
UC_DLBCL 449 Validation 10.01 5.37 12.17 -6.65
UC_DLBCL1458 Training 7.50 5.79 9.60 -5.40
UC DLBCL_460 Validation , 10.26 8.27 12.29 -7.27
UC DLBCL_491 Training 9.43 4.73 12.39 -6.40
UC_ DLBCL 528 Validation 8.42 6.19 11.63 -6.18
UC_DLBCL1615 Validation 8.44 9.01 12.80 -6.92
UC DLBCL 625 Training 10.43 8.27 12.62 -7.39
UC_DLBCL_664 Training 9.80 8.74 12.72 -7.29
UC_DLBCL 671 Training 9.42 5.26 11.53 -6.32
UC_DLBCL1682 Training 9.01 4.73 12.33 -6.26
UC DLBCL_683 Training 8.85 8.23 12.57 -6.87
UC _ DLBCL_ 684 Validation 9.62 8.78 12.76 -7.25
UC DLBCL 748 Validation _ 7.60 5.79 9.55 -5.42
UC_DLBCL_751 Training 6.40 9.91 13.14 -6.50
UC_DLBCL_808 Training 9.44 7.01 13.09 -6.95
_
UC DLBCL 831 Validation 9.45 5.81 11.58 -6.43
UC_DLBCL_834 Training 8.52 7.66 11.77 -6.50
UC _ DLBCL_ 838 Validation 8.49 4.60 12.56 -6.11
UC DLBCL_851 Validation 7.50 4.82 8.19 -4.94
UC_ DLBCL_ 854 Validation 8.35 5.82 12.59 -6.29
UC_DLBCL_855 Training 9.56 5.44 12.08 -6.51
UC_DLBCL_856 Validation 6.81 7.49 9.32 -5.42
[00167] In order to visualize the predictive power of the model, the 200
samples
were ranked according to their survival predictor scores and divided into four
104
CA 2897828 2019-12-20

quartiles. Kaplan-Meier plots of overall survival probability show clear
differences in
survival rate between these four quartiles (Figure 12).
Example 9: Development of a refined DLBCL survival predictor based on qenomic
alterations:
[00168] Comparative genomic hybridization (CGH) was performed on 224 DLBCL
samples for which gene expression profiles had previously been obtained using
a
Lymphochip (Rosenwald 2002). Of these 224 samples, 87 had been classified as
GCB based on the Lymphochip expression data, 77 had been classified as ABC, 19
had been classified as PMBL, and 41 were unclassified (Wright 2003). Clinical
data
was available for each subject from whom the samples had been obtained
(Rosenwald 2002). All patients had received anthracycline-based chemotherapy.
Median follow-up was 2.7 years and 58% of patients died during this period.
The
median age of the patients was 60 years and 54% were men. 16% of patients had
Ann Arbor stage I disease and 30%, 19%, and 35% had Stage II, Ill, and IV,
respectively. 38% of DLBCL patients (78 cases) with available data were in the
low-
risk IPI group (IPI 0-1), 48% (99 cases) were in the intermediate-risk IPI
group (IPI 2-
3), and 14% (30 cases) were in the high-risk IPI group (IPI 4-5).
[00169] CGH was carried out using a commercially available kit (Vysis,
Downers Grove, IL). Hybridization and digital image acquisition, processing,
and evaluation were performed on a Cytovision Ultra workstation (Applied
Imaging, Sunderland, UK) as described previously (Bea 1999). Signal ratios
greater than 1.25 were considered chromosomal gains, while signal ratios less
than 0.75 were considered chromosomal losses. Ratios exceeding 1.5 and/or
strong focal signals with a ratio profile showing over-representation were
considered genomic amplifications.
105
CA 2897828 2019-12-20

[00170] CGH alterations in individual cytobands were treated as categorical
variables and their associations with DLBCL subgroups or gene expression
signatures were analyzed as follows. Preliminary analyses did not reveal
significant
differences in the effects of gains and amplifications, so they were treated
as
equivalent chromosomal abnormalities. Since a large number of individual
chromosomal abnormalities were analyzed, there was a danger that some of the
abnormalities would appear to be significant purely by chance. To avoid such
false
positives, a stepwise permutation test that generated nominal p-values
accounting
for multiple hypothesis testing was used (Westfall 1993; Simon 2003). This
test
takes into account the correlation between different chromosomal
abnormalities.
Differences in abnormality frequency between subtypes were detected using a
chi
squared test. Differences in gene expression signature measures affected by
genomic imbalances were detected using a t-test. To further reduce the effects
of
multiple comparisons, only those chromosomal abnormalities that were present
in a
substantial portion of the data were analyzed. For the subgroup analysis, only
those
alterations that had a frequency of >20% in one or more of the DLBCL subgroups
were considered. For correlation with gene expression signatures, chromosomal
abnormalities were only considered if they occurred in at least 5% of all
DLBCL
samples. P-values for the association between gene expression levels (as a
continuous variable) and genomic imbalances (amplification vs. gain vs. normal
copy
number) were calculated using an ANOVA test. P-values of <0.01 were considered
significant to account for multiple comparisons. Overall survival was modeled
using
a Cox proportional hazards approach and visualized using the Kaplan-Meier
method. The P values were adjusted for multiple comparisons, with the follow-
up
time and status at follow-up being permuted. Once an abnormality was found to
be
106
CA 2897828 2019-12-20

significant univariately, a likelihood ratio test was used to determine
whether this
variable added significantly to the survival model based on gene expression.
[00171] Figure 29 shows the results of CGH analysis for GCB (A), ABC (B), and
PMBL (C) samples (Bea 2005). Overall, chromosomal alterations were observed in
164 of the 224 patients (73%). A summary of the most common alterations is set
forth in Table 2415.
Table 2415:
Overall ABC GCB PMBL Unclassified
(n=224) (n=77) (n=87) (n=19) (n=41)
Samples exhibiting 164 (73%) 63 (81%) 63 (72%) 16 (84%) 22
(54%)
alterations
Mean number of 3.3 4.5 3.1 3.3 1.7
alterations
Mean number of gains 1.9 2.5 1.6 2.1 1.0
Mean number of 0.3 0.4 0.3 0.4 0.0
amplifications
Mean number of losses 1.2 1.6 1.1 0.8 0.6
Gains
Xp 27(12%) 12(16%) 12(14%) 3(16%)
0
1q25-q32 26 (12%) 9 (12%) 9(10%) 1(5%) 7
(17%)
2p14-p16* 39(17%) 12(15%) , 15(17%) ,
9(47%) 3(7%)
Trisomy 3* 14 (6%) 12 (15%) 0 1 (5%) 1
(2%)
3p+ 28 (12%) 24 (31%) 1 (1%) 1(5%) 2
(5%)
3q* 22 (10%) 20 (26%) 0 1(5%) 1(2%)
3q27-qter* 35 (16%) 26 (33%) 4 (5%) 3 (16%)
2 (5%)
6p 30 (13%) 13 (17%) 11(13%) 1(5%)
5(12%)
7p 22 (10%) 8 (10%) 13 (15%) 1(5%)
0
7q 25(11%) 10(13%) 13 (15%) 1(5%)
, 1(2%)
8q23-qter 23(10%) 8(10%) 10(11%) 2(11%)
3(7%)
9p* 14 (6%) 5 (6%) 0 7 (37%) 2
(5%)
12p , 19 (8%) 4 (5%) , 14 (16%) 1(5%) 0
12q12# 24(11%) 4(5%) 18 (21%) 1(5%) 1(2%)
12q22-qter 22 (10%) 7 (9%) 13 (15%) 1(5%) 1(2%)
18q21-q22+ 42 (19%) 26 (34%) 9 (10%) 3(16%)
4 (10%)
Losses
6q16 50 (22%) 26 (34%) 19 (22%) 0
5(12%)
6q21-q22* 55 (25%) 31(40%) 19 (22%) 0
5 (12%)
8p22-pter 19 (8%) 8 (10%) 3 (3%) 3 (16%) 5
(12%)
17p 22 (10%) 14 (18%) 7 (8%) 0
1(2%)
" P<0.001; + P<0.05; # P=0.059; unclassified tumors were not included in
statistical analysis
[00172] The number of alterations did not differ statistically between GCB
(3.1 3.7,
n=87), ABC (4.5 4.5, n=77), PMBL (3.3 2.7, n=19) and unclassified DLBCL
(1.7 2.2, n=41). Among samples exhibiting alterations, 81% exhibited more than
one. The most frequent alteration in those samples exhibiting only a single
107
CA 2897828 2019-12-20

alteration was loss of 6q (n=8), with two minimally lost regions in 6q21-q22
and
6q25-qter. These deletions may represent early events in the development of
these
lymphomas.
[00173] Irrespective of the DLBCL subgroup, the most frequent alterations were
loss
of 6q22-q22 (25%), loss of 6q16 (22%), gain of 18q21-q22 (19%), gain of 2p14-
p16
(17%), gain of 3q27-qter (16%), gain of 6p (13%), and gain of Xp, 1q25-q32,
and 3p
(12% each). Amplifications were identified in 33 different chromosomal
regions,
most frequently in 2p14-p16 and 18q21-q22 (11 and 20 cases, respectively).
Some
alterations occurred frequently in the same tumors, suggesting that they may
be part
of a recurrent lymphogenesis pathway. For example, 17 of 26 ABC samples with
3q27-qter gains also exhibited 18q21-q22 gains (P=0.0001, odds ratio: 9.23;
95% Cl
3.14-27.2).
[00174] Notably, several chromosomal alterations were differentially
distributed
among DLBCL subgroups (Figure 290). ABC exhibited characteristic and recurrent
gains of chromosome 3, gains and amplification of 18q21-q22, and loss of 6q21-
q22. Gains of the whole 3q arm and trisomy 3, which were observed in 26% and
15% of ABC samples, respectively, were never observed in GCB and observed only
once in PMBL. Gains of 18q21-q22 were observed in 34% of ABC samples, versus
only 10% and 16% of GCB and PMBL samples, respectively. Amplification of
18q21, which contains the BCL2 gene, was also more frequent in ABC (18%) than
in
GCB or PMBL (5% each). Previous studies utilizing PCR-based and FISH methods
found that the t(14;18) translocation, which involves the BCL2 gene, occurs in
GCB
but never in ABC (Huang 2002; lqbal 2004). Interestingly, 3 of the 4 GCB
samples
exhibiting amplification of 18q21 also exhibited the t(14;18) translocation
(the
remaining case was not analyzed for this translocation). High expression of
the
108
CA 2897828 2019-12-20

BCL2 gene is a characteristic feature of ABC, but only occurs in GCB that have
the
t(14;18) translocation (Alizadeh 2000; Huang 2002; Rosenwald 2002). Together,
these data suggest that amplification of the 18q21 region occurs
preferentially in
lymphomas that have the ability to express the BCL2 gene.
[00175] GCB samples were characterized by more frequent gains of 12q12
compared to ABC and PMBL, although this increase did not reach statistical
significance (21% for GCB vs. 5% for both ABC and PMBL, P=0.059). PMBL
samples were characterized by frequent gains of 9p21-pter (37% for PMBL vs. 0%
for GCB and 6% for ABC, P<0.001) and 2p14-p16 (47% for PMBL vs. 17% for GCB
and 11% for ABC, P<0.02) compared to GCB and ABC. Taken together, these data
further demonstrate that GCB, ABC, and PMBL are genetically distinct.
[00176] To confirm some of the more frequent DLBCL chromosomal alterations
identified by CGH, real-time quantitative PCR (RQ-PCR) was used to quantify
the
copy number of select genes from the following regions: 2p14-16 (REL, BCL11A),
12q13-q14 (SAS, CDK4, MDM2), 3q27 (RFC4, BCL6, and 18q21 (MADH4, MALT1,
BCL2). RQ-PCR was performed using the ABI PrismTM 7700 Sequence Detector
System (Applied Biosystems). 132-microglobulin (J32M) was used as a reference
gene. Each assay was analyzed using the comparative cycle threshold (CT)
method,
using the arithmetic formula provided by the manufacturer. To determine the
cut-off
values for a genomic gain/amplification in each probe set, eight DNA samples
from
peripheral blood or placenta of healthy subjects was studied. The cut-off
value for a
genomic gain was determined to be the mean ratio plus three standard deviation
units (approximately 1.3 for each gene). A ratio between the cut-off value and
2 was
considered a gain, while a ratio of greater than 2 was considered an
amplification. A
subset of samples were also investigated using albumin (ALB) as a reference
gene.
109
CA 2897828 2019-12-20

The results between the two control genes were totally concordant in 87% of
the
cases, and partially concordant in 13% of the cases. For the four samples that
CGH
had shown contained alterations in the /32M locus (15q21.1), ALB was used as
the
sole reference gene.
[00177] REL was found to be amplified in virtually all GCB samples in which
high-
level 2p14-16 amplifications had been observed by CGH. The copy number of
BCL11A was increased in all but one of these cases, albeit usually at lower
levels
than REL. Although CGH had shown high-level 2p14-16 amplifications in two ABC
samples, RQ-PCR showed merely a gain in REL for these two samples. BCLIIA
was amplified in one of these samples and gained in the other. These results
confirm the previous observation that REL may not be the primary target of
amplification in ABC (Rosenwald 2002). CDK4 and SAS, which map to 12q13-q14,
were frequently gained or amplified in GCB samples that had exhibited 12q
gains by
CGH, whereas MDM2 was less commonly altered in these samples. In contrast, all
three ABC samples with 12q13-q14 gains exhibited gains of CDK4, although less
frequently than gains of SAS or MDM2. RFC4 and BCL6, located in 3q27, were
gained or amplified in all ABC and GCB samples in which 3q27-qter gains or
amplifications had been observed by CGH. MALT1 and BCL2 were commonly
gained or amplified in ABC and GCB samples that had exhibited 18q21 gains by
CGH, while MADH4 was less frequently altered.
[00178] To determine the influence of these chromosomal alterations on locus-
specific gene expression, mRNA levels of genes located in four recurrently
gained/amplified regions (2p14-p16, 3q27-qter, 12q12-q15, and 18q21-q22) were
correlated with copy number changes. It was found that each of these
chromosomal
110
CA 2897828 2019-12-20

alterations was associated with a higher expression levels for a subset of
genes
within the region, but these genes differed between DLBCL subgroups.
[00179] 14 genes were mapped to the chromosomal region 2p14-p16. GCB and
ABC samples with increased genomic copy numbers in this region showed
significant overexpression of 8 (57%) and 5 (36%) of these 14 genes,
respectively
(Figure 30A and 30B). Four of the genes (VRK2, XP01, SLC14A, and ACTR2)
were significantly overexpressed in both GCB and ABC samples (Figure 30A and
2B). In contrast, REL, ASHA2, MDH1, and UGP2 were only overexpressed in GCB-
DLBCL with 2p14-p16 gains (Figure 30A).
[00180] GCB and ABC samples with gains/amplifications in the 12q12-q15 region
showed significant overexpression of 10(19%) and 12(23%) of the 52 genes
represented on the Lymphochip microarray. Five of these genes were
overexpressed in both GCB and ABC samples (SENP1, MCRS1, MARS, SAS, and
CDK4) (Figure 30F). Most of the overexpressed genes clustered to the
chromosomal region 12q13.
[00181] 7 (33%) of the 21 genes mapping to chromosome 3q27-qter were
significantly overexpressed in ABC (Figure 30C), versus only 2 (13%) in GCB.
Similarly, 9(75%) of the 12 genes mapping to chromosome 18q21-q22 were
significantly overexpressed in ABC (Figure 30E), versus only 4 (33%) in GCB
(Figure
30D). All 4 18q21-q22 genes overexpressed in GCB (MADH2, MADH4, L0051320,
and PMAIP1) were also overexpressed in ABC.
[00182] To determine whether the various genetic alterations identified in
DLBCL
samples influence previously defined gene expression signatures, gene
expression
signature averages were created for each DLBCL, and then evaluated within
tumors
with specific chromosomal alterations (Bea 2005). Statistically significant
111
CA 2897828 2019-12-20

associations were observed between several chromosomal alterations and the
proliferation, lymph node, 1-cell, and MHC class II gene expression
signatures. In
particular, gains of various cytobands of chromosome 3 and losses in 6q21 were
both associated with increased expression of the proliferation gene expression
signature (Figure 31). Copy number gains of the chromosomal regions 3p12 and
3q12 were associated with decreased expression of the MHC class II gene
expression signature. Genetic losses of 6q21 and other cytobands of chromosome
6, as well as losses of 17p13, gains of Xp11, gains of 11q24-q25, gains of
12q12,
and gains of several cytobands in 7p and 7q all decreased expression of the 1-
cell
gene expression signature. Finally, gains of Xp21 were associated with
increased
expression of the lymph node gene expression signature, while gains of 3q22 or
several additional cytobands in 3p and 3q were associated with decreased
expression of lymph node gene expression signature.
[00183] The prognostic value of each DLBCL chromosomal alteration was analyzed
across all DLBCL samples and within each DLBCL subtype separately. Although
several chromosomal alterations were individually associated with a
significant
increase or decrease in overall survival rate, only gains within certain
regions of
chromosome 3 were significantly associated with shorter overall survival after
adjustment for multiple comparisons in the whole series of patients. These
regions
were 3p11-p12, 3q11-q13, 3q21-q24, and 3q25-q27.
[00184] Previously, a gene expression-based DLBCL survival predictor had been
developed using four gene expression signatures and BMP6 (Rosenwald 2002).
This survival predictor had the following formula:
Survival predictor score = [0.241*(proliferation gene expression
signature value)] + [0.310*(BMP6)] ¨ [0.290*(germinal center B cell
112
CA 2897828 2019-12-20

gene expression signature value)] ¨ [0.311*(MHC class ll gene
expression signature value)] ¨ [0.249*(lymph node gene expression
signature value)].
[00185] This model could divide DLBCL patients into four quartile groups with
5-year
survival rates of 73%, 71%, 34%, and 15%. To determine whether the chromosome
3 gains discussed above could improve this survival predictor, a multivariate
analysis
was performed. In this analysis, chromosome 3 gains involving the 3p11-p12
region
had an independent prognostic value and improved survival predictions obtained
using gene expression data alone (Figure 32). Cases with gains of 3p11-p12
were
primarily those that had been categorized in the least favorable quartile
survival
group using gene expression data alone. However, several cases with 3p11-p12
gains had been included in the more favorable quartile survival groups based
on
gene expression data. These cases had a significantly worse clinical prognosis
than
would have been predicted by gene expression data alone.
Example 10: Development of an MCL survival predictor using gene expression
data
from Affymetrix U133A and U133B microarrays:
[00186] The connection between higher expression of proliferation genes and
worse
survival in MCL had previously been documented and validated (Rosenwald
2003a).
A cluster of proliferation genes had been identified in the DLBCL samples used
to
create the DLBCL survival predictor described in Example 7. By averaging the
expression of these genes, a proliferation gene expression signature value had
been
developed for the DLBCL samples. The correlation of this signature with each
probe
set on the U133A and U133B microarrays was determined, and the 22 genes for
which the correlation was greater than 0.5 were labeled proliferation genes.
The
correlation between expression of these proliferation genes and survival in 21
MCL
113
CA 2897828 2019-12-20

samples was estimated using the Cox proportional hazards model. Table 2377
lists
these 21 MCL samples.
114
CA 2897828 2019-12-20

Table 2377
Sample ID # Length of follow-up Status at follow-up
Used in creating
(years) survival predictor?
MCL 1012 3.19 Alive Yes
MCL_1091 3.03 Alive Yes
MCL_1114 0.59 Dead Yes
MCL_1128 0.43 Dead Yes
MCL 1150 3.21 Dead Yes
MCL11162 0.78 Alive Yes
MCL_1166 0.53 Dead Yes
MCL 1194 0.55 Alive Yes
MCL _885 1.19 Alive Yes
MCL 918 1.95 Dead Yes
MCL 924 5.48 Dead Yes
MCL_925 7.23 Alive Yes
MCL_926 5.18 Dead Yes
MCL 936 2.80 Alive Yes
MCL¨_939 1.07 Dead Yes
MCL_953 2.31 Dead Yes
MCL_956 1.40 Dead Yes
MCL 964 0.75 Alive Yes
MCL_966 0.21 Dead Yes
MCL_968 1.59 Dead Yes
MCL_970 5.02 Dead Yes
Out of the 22 proliferation genes, 11 were significant at a 0.001 level. The
expression level of these 11 genes in each of the 21 MCL samples was averaged
to
generate a proliferation gene expression signature value. No other genes
represented on the U133A or U133B microarrays correlated with MCL survival to
an
extent greater than would be expected by chance, so the final model included
only .
proliferation genes. The 11 genes used to generate the model are presented in
Table 2378.
Table 2378
Signature UNIQID Gene Symbol
Proliferation 1097290 CIRH1A
Proliferation 1101295 FLJ40629
Proliferation 1119729 TK1
Proliferation 1120153 LMNB1
Proliferation 1120494 CDC6
Proliferation 1124745 KIAA0056
Proliferation 1126148 DKFZp586E1120 .
Proliferation 1130618 TPIl
Proliferation 1134753 WHSC1
Proliferation 1139654 ECT2
Proliferation 1140632 IMAGE:52707
115
CA 2897828 2019-12-20

[00187]A survival predictor score for MCL was generated using the following
equation:
Survival predictor score = 1.66*(proliferation gene expression signature
value).
This model was associated with survival in a statistically significant manner
(p =
0.00018). To illustrate the significance of the model in predicting survival,
the 21
MCL samples were divided into two equivalent groups based on their survival
predictor scores. Those samples with survival predictor scores above the
median
were placed in the high proliferation group, while those with survival
predictor scores
below the median were placed in the low proliferation group. Figure 13
illustrates
the Kaplan Meier survival estimates for these two groups. Median survival for
the
high proliferation group was 1.07 years, while median survival for the low
proliferation group was 5.18 years.
Example 11: Development of an MCL survival predictor using gene expression
data
from the Lymph Dx microarray:
[00188]A set of 21 genes associated with proliferation and poor prognosis in
MCL
had been identified previously (Rosenwald 2003a). Of these 21 genes, only four
were represented on the Lymph Dx microarray. In order to find a larger set of
genes
on the Lymph Dx microarray associated with survival in MCL, Lymphochip
expression data (Rosenwald 2003a) was re-analyzed and another set of
proliferation
genes whose expression levels were correlated with poor survival in MCL were
identified. Thirteen of these genes were represented on the Lymph Dx
microarray
(median expression >6 on 10g2 scale). These 13 genes are listed in Table 2379.
Table 2379
Signature UNIQID Unigene ID Build 167 Gene symbol
Proliferation 1119294 156346 TOP2A
Proliferation 1119729 164457 TK1
116
CA 2897828 2019-12-20

=
Proliferation 1120153 89497 LMNB1
Proliferation 1121276 24529 CHEK1
Proliferation 1123358 442658 AURKB
Proliferation 1124178 446579 HSPCA
Proliferation 1124563 249441 WEE1
Proliferation 1130799 233952 PSMA7
Proliferation 1131274 374378 CKS1B
Proliferation 1131778 396393 UBE2S
Proliferation 1132449 250822 STK6
Proliferation 1135229 367676 DUT
Proliferation 1136585 80976 MKI67
[00189] The expression levels of the 13 genes listed in Table 2379 on the
Lymph Dx
microarray were transformed into the 10g2 scale and averaged to form a
proliferation
gene expression signature value. This was used to generate a survival
predictor
score using the following equation:
Survival predictor score = 1.66*(proliferation gene expression signature
value).
[00190] For the 21 MCL samples analyzed, the survival predictor score had a
mean
of 14.85 and a standard deviation of 1.13. Even in this limited sample set,
the
survival predictor score was significantly associated with prognosis
(p=0.0049), with
each unit increase in the score corresponding to a 2.7 fold increase in the
relative
risk of death. Data for all 21 samples is shown in Table 2380.
Table 2380
Sample ID # Proliferation Survival predictor
signature value score
MCL 1012 8.83 14.658
MCL 1091 8.81 14.625
MCL 1114 10.39 17.247
MCL_1128 10.12 16.799
MCL 1150 8.33 13.828
MCL 1162 8.15 13.529
MCL_1166 9.40 15.604
MCL 1194 7.44 12.350
MCL_885 8.68 14.409
MCL_918 9.33 15.488
MCL_924 8.35 13.861
MCL 925 8.86 14.708
MCL_926 8.14 13.512
MCL 936 8.56 14.21
MC L:939 9.14 15.172
MCL_953 9.25 15.355
117
CA 2897828 2019-12-20

MCL 956 9.35 15.521
MCL 964 9.74 16.168
MCL_966 8.76 14.542
MCL_968 9.10 15.106
MCL_970 9.27 15.388
[00191] To illustrate the significance of the model in predicting survival,
the 21 MCL
samples were divided into two equivalent groups based on their survival
predictor
scores. Those samples with survival predictor scores above the median were
placed in the high proliferation group, while those with survival predictor
scores
below the median were placed in the low proliferation group. Figure 14
illustrates
the Kaplan Meier survival estimates for these two groups.
Example 12: Identification of lymphoma samples as MCL based on Bayesian
analysis of gene expression data from Affymetrix U133A and U133B microarrays:
(00192]A statistical method based on Bayesian analysis was developed to
distinguish MCL samples from samples belonging to other lymphoma types based
on gene expression profiling. This method was developed using the gene
expression data obtained in Example 1 for the following lymphoma types: ABC,
GCB, PMBL, BL, FH, FL, MALT, MCL, PTLD, SLL, and splenic marginal zone
lymphoma (splenic). Tables 1707-1741 (discussed in Example 1) provide gene
expression data for samples within each of these lymphoma types, including the
expression level of each gene and the difference in expression of each gene
between types. Tables 1710, 1715, and 1723 (corresponding to FL, MCL, and
DLBCL, respectively) include the correlation between expression of each gene
and
survival.
[00193] To determine the lymphoma type of a sample, a series of predictor
models
are generated. Each predictor model calculates the probability that the sample
belongs to a first lymphoma type rather than a second lymphoma type. A method
118
CA 2897828 2019-12-20

was developed to determine whether a sample was MCL, or one of the following
lymphoma types: ABC, BL, FH, FL, GCB, MALT, PMBL, PTLD, SLL, or splenic.
This method required ten different predictor models, each designed to
determine
whether the sample belonged to MCL or one of the other ten lymphoma types
(e.g.,
MCL vs. ABC, MCL vs. BL, etc.).
[00194] Several of the lymphoma samples analyzed displayed a tendency towards
elevated or reduced expression of genes from the lymph node and proliferation
gene
expression signatures. These genes are likely to be highly differentially
expressed
between the lymphoma types, but they do not serve as good predictor genes
because they are often variably expressed within a single lymphoma type. For
this
reason, any gene that displayed a correlation with the proliferation or lymph
node
signatures was eliminated from consideration.
[00195] For each lymphoma type pair (e.g., MCL vs. ABC, MCL vs. FL, etc.), 20
genes were identified that exhibited the greatest difference in expression
between
MCL and the second lymphoma type according to a Student's t-test. The choice
to
use 20 genes was arbitrary. For each sample X, the 20 genes were used to
generate a linear predictor score (LPS) according to the following formula:
LPS(X)= Et,x),
where Xj is the expression of gene) in sample X and tj is the t-statistic for
the
difference in expression of gene] between a first lymphoma type and a second
lymphoma type. This is merely one method for generating an LPS. Others methods
include linear discriminant analysis (Dudoit 2002), support vector machines
(Furey
2000), or shrunken centroids (Tibshirani 2002). In addition, there is no
requirement
that a t-statistic be used as the scaling factor.
119
CA 2897828 2019-12-20

[00196]After an LPS had been formulated for each lymphoma sample, the mean
and standard deviation of these LPS's was calculated for each lymphoma type.
For
a new sample X, Bayes' rule can be used to estimate the probability that the
sample
belongs to a first lymphoma type rather than a second lymphoma type (Figure
15).
In this example, Bayes' rule was used to calculate the probability q that
sample X
was MCL rather than a second lymphoma type using the following equation:
q(X is type 1) =
eft(LPS(X); J1,o ) + 0(LPS(X); p2 ,a2)
where type us MCL, type 2 is one of the other nine lymphoma types, 0(x; ,a)
is
the normal density function with mean and standard deviation o- , fi and 6,
are the
sample mean and variance of the LPS values for lymphoma type 1, and ando-,
are the sample mean and variance of the LPS values for lymphoma type 2.
[00197] This method was used to develop ten predictor models, one for each
pairing
of MCL and a second lymphoma type. A sample was classified as MCL if each of
the ten predictors generated at least a 90% probability that the sample was
MCL. If
any of the ten predictors indicated a probability of less than 90%, the sample
was
classified as non-MCL.
[00198] The 10 sets of 20 genes that were included in these models and the t-
statistics for each gene are presented in Tables 2381-2490.
Table 2381: MCL vs. ABC predictor genes
UNIQID Gene name Scale Factor
1103711 Homo sapiens cDNA FLJ11833 fis, clone HEMBA1006579.
17.88496416
1133111 PDE9A ¨ phosphodiesterase 9A 17.61579873
1137987 PLXNB1 plexin B1 17.47030156
1132835 SOX11 SRY (sex determining region Y)-box 11 16.89404131
Homo sapiens, Similar to L0C168058, clone MGC:39372
1109505 IMAGE:5089466, mRNA, complete cds 15.78111902
L0058486 transposon-derived Buster1 transposase-like
1139054 protein 15.77800815
TIA1 TIA1 cytotoxic granule-associated RNA binding
1119361 protein 15.68070962
1115226 K1AA1683 KIAA1683 protein 15.67954057
120
CA 2897828 2019-12-20

1101211 Homo sapiens cDNA: FLJ21960 fis, clone HEP05517. 15.4183527
1118963 Homo sapiens cDNA FLJ35653 fis, clone SPLEN2013690.
15.36802586
1096503 GL012 -- hypothetical protein GL012 14.64776335
1127849 SNN stannin 14.54859775
Homo sapiens mRNA; cDNA DKFZp586K1922 (from clone
1099204 DKFZp586K1922) 14.32724822
1098840 C3orf6 -- chromosome 3 open reading frame 6 14.10346944
1139444 RABL2B RAB, member of RAS oncogene family-like 2B
14.10016196
1106855 K1AA1909 KIAA 1909 protein 13.9504946
1126695 K1AA0484 KIAA0484 protein 13.92285415
1120137 , FCGBP Fc fragment of IgG binding protein 13.86147896
1133011 TMSNB thymosin, beta, identified in neuroblastoma cells
13.74377784
1133192 GRP3 -- guanine nucleotide exchange factor for Rap1 -
17.09085725
Table 2382: MCL vs. BL predictor genes
UNIQID Gene name Scale Factor
1120900 EPHB6 EphB6 13.43582327
1112061 Homo sapiens cDNA FLJ90513 fis, clone NT2RP3004355.
12.73065392
Homo sapiens, Similar to L0C168058, clone MGC:39372
1109505 IMAGE:5089466, mRNA, complete cds 12.63674985
1133099 DNASE1L3 deoxyribonuclease I-like 3 12.43333984
1106855 KIAA1909 K1AA1909 protein 12.32623489
1110070 ESTs 12.05416064
1121739 ZNF135 -- zinc finger protein 135 (clone pHZ-17) 11.90460363
1098840 C3orf6 -- chromosome 3 open reading frame 6 11.90309143
1132833 SOX11 SRY (sex determining region Y)-box 11 11.60864812
1121693 KIAA0450 KIAA0450 gene product 11.33634052
ILT7 -- leukocyte immunoglobulin-like receptor, subfamily A
1123760 (without TM domain), member 4 11.18744726
1125964 K1AA0792 K1AA0792 gene product 11.14762675
1112306 ESTs 11.02434114
1096070 DNMT3A -- DNA (cytosine-5-)-methyltransferase 3 alpha
10.98991879
Homo sapiens, similar to Zinc finger protein 85 (Zinc finger
1129943 protein HPF4) (HTF1), clone IMAGE:3352451, mRNA 10.72494956
1118749 PRKWNK1 --protein kinase, lysine deficient 1 10.64623382
1098954 FLJ 13204 -- hypothetical protein FLJ13204 10.46164401
1134749 PRKCBP1 --protein kinase C binding protein 1 10.40948157
1131860 BIN1 --bridging integrator 1 10.31084561
TGFBR2 -- transforming growth factor, beta receptor II
1123148 (70/80kDa) 10.2956213
Table 2383: MCL vs. FH predictor genes
UNIQID Gene name Scale Factor
1132834 SOX11 SRY (sex determining region Y)-box 11 24.3531072
1100873 ESTs 16.83342764
1109603 ESTs 13.02401995
1139411 OSBPL10 oxysterol binding protein-like 10 12.54369577
1106855 K1AA1909 KIAA1909 protein 12.10316361
1125193 CNR1 cannabinoid receptor 1 (brain) 12.070579
1137450 ALOX5 arachidonate 5-lipoxygenase 11.74571823
121
CA 2897828 2019-12-20

1100258 K1AA1384 KIAA1384 protein 11.60998697
1133167 ZFD25 -- zinc finger protein (ZFD25) 11.52931491
PPFIBP2 PTPRF interacting protein, binding protein 2
1136831 (liprin beta 2) 11.50062692
1138222 NA 10.99674674
Homo sapiens mRNA; cDNA DKFZp66761913 (from clone
1099437 DKFZp667B1913) 10.90797288
SPAP1 SH2 domain containing phosphatase anchor
1140236 protein 1 10.77082801
1114109 DCAL1 dendritic cell-associated lectin-1 10.65867119
1098277 PRICKLE1 -- prickle-like 1 (Drosophila) 10.55457068
CD24 CD24 antigen (small cell lung carcinoma cluster 4
1135138 antigen) 10.41999962
1103304 Homo sapiens clone CDABP0095 mRNA sequence -10.46625233
1128460 RDGBB --retinal degeneration B beta -10.91106245
1121953 KIAA0125 KIAA0125 gene product -11.22466255
1129281 C14orf110 --chromosome 14 open reading frame 110 -
15.54465448
Table 2384: MCL vs. FL predictor genes
UNIQID Gene name Scale Factor
1132835 SOX11 SRY (sex determining region Y)-box 11 22.14208817
1096070 DNMT3A -- DNA (cytosine-5-)-methyltransferase 3 alpha
20.53740132
1103711 Homo sapiens cDNA FLJ11833 fis, clone HEMBA1006579.
20.49880004
1137987 PLXNB1 plexin B1 18.38081568
Homo sapiens, Similar to L0C168058, clone MGC:39372
1109505 IMAGE:5089466, mRNA, complete cds 17.17812448
1098840 C3orf6 -- chromosome 3 open reading frame 6 16.32703666
1130926 C5orf13 -- chromosome 5 open reading frame 13 15.34261878
1096396 SPG3A -- spastic paraplegia 3A (autosomal dominant)
14.75437736
1132734 COL9A3 -- collagen, type IX, alpha 3 14.684583
1139393 OPN3 opsin 3 (encephalopsin, panopsin) 14.39118445
1115537 L0084518 -- protein related with psoriasis 14.18446144
1102215 Homo sapiens cDNA FLJ11666 fis, clone HEMBA1004672.
14.16246426
Homo sapiens cDNA: FLJ21930 fis, clone HEP04301,
highly similar to HSU90916 Human clone 23815 mRNA
1124585 sequence. -14.33315955
1137561 HOXA1 homeo box A1 -15.38404642
Homo sapiens mRNA; cDNA DKFZp667A1115 (from clone
1100581 DKFZp667A1115) -15.91666634
1124646 KIAA0084 KIAA0084 protein -16.40577696
1114543 ESTs -17.60167863
1120090 BCL6--B-cell CLL/Iymphoma 6 (zinc finger protein 51) -
17.63091181
1123731 RGS13 -- regulator of G-protein signalling 13 -22.41602151
1133192 GRP3 -- guanine nucleotide exchange factor for Rap1 -
27.28308723
Table 2385: MCL vs. GCB predictor genes
UNIQID Gene name Scale Factor
1098840 C3orf6 -- chromosome 3 open reading frame 6 22.26488562
1132835 SOX11 SRY (sex determining region Y)-box 11 17.76179754
1137987 PLXNB1 plexin B1 16.86845147
1098954 FLJ13204 -- hypothetical protein FLJ13204 16.65023669
122
CA 2897828 2019-12-20

1103711 Homo sapiens cDNA FLJ11833 fis, clone HEMBA1006579.
15.64719784
1096070 DNMT3A -- DNA (cytosine-5-)-methyltransferase 3 alpha
15.22540494
1139393 OPN3 opsin 3 (encephalopsin, panopsin) 14.64030565
1127849 SNN stannin 14.28242206
Human HeLa mRNA isolated as a false positive in a two-
1098156 hybrid-screen. 14.00049272
1128845 FLJ20174 -- hypothetical protein FLJ20174 13.96064416
Homo sapiens, similar to Zinc finger protein 85 (Zinc finger
1129943 protein HPF4) (HTF1), clone IMAGE:3352451, mRNA 13.85404507
1140116 DKFZP564B116 --hypothetical protein DKFZp564B1162
13.81464172
1106855 K1AA1909 K1AA1909 protein 13.74521849
1120900 EPHB6 EphB6 13.46567004
1127371 Homo sapiens cDNA FLJ14046 fis, clone HEMBA1006461.
13.45735668
TIA1 TIA1 cytotoxic granule-associated RNA binding
1119361 protein 13.37376559
EDG1 -- endothelial differentiation, sphingolipid G-protein-
1120854 coupled receptor, 1 13.1047657
1098277 PRICKLE1 -- prickle-like 1 (Drosophila) 13.04993076
,
1140127 TRIM34 -- tripartite motif-containing 34 12.66260609
Homo sapiens mRNA; cDNA DKFZp667A1115 (from clone
1100581 DKFZp667A1115) -12.81251689
Table 2386: MCL vs. MALT predictor genes
UNIQID Gene name Scale Factor
1132834 SOX11 SRY (sex determining region Y)-box 11 20.7489202
1101987 KIAA1909 KIAA1909 protein 10.78991326
1100873 ESTs 10.11845036
1130764 HNRPAO -- heterogeneous nuclear ribonucleoprotein AO
9.432459453
Homo sapiens, Similar to thymosin, beta, identified in
neuroblastoma cells, clone MGC:39900 IMAGE:5247537,
1102178 mRNA, complete cds 9.035605572
1098277 PRICKLE1 -- prickle-like 1 (Drosophila) 9.003360784
1130926 C5orf13 -- chromosome 5 open reading frame 13 8.712830747
1098694 LOCI 12868 -- hypothetical protein LOC112868 8.309789856
1103711 Homo sapiens cDNA FLJ11833 fis, clone HEMBA1006579.
8.248526605
1138099 NA 8.107440225
EDG1 -- endothelial differentiation, sphingolipid G-protein-
1120854 coupled receptor, 1 8.045872672
1102215 Homo sapiens cDNA FLJ11666 fis, clone HEMBA1004672.
8.032351578
1121739 ZNF135 --zinc finger protein 135 (clone pHZ-17) 8.020919565
1096070 DNMT3A -- DNA (cytosine-5-)-methyltransferase 3 alpha
7.964477216
1101211 Homo sapiens cDNA: FLJ21960 fis, clone HEP05517. 7.738742472
CHL1 -- cell adhesion molecule with homology to L1 CAM
1120825 (close homolog of L1) 7.516130116
Homo sapiens mRNA; cDNA DKFZp667B1913 (from clone
1099437 DKFZp6671:31913) 7.209041652
1096503 GL012 -- hypothetical protein GL012 7.171540413
LILRA2 -- leukocyte immunoglobulin-like receptor,
1135927 subfamily A (with TM domain), member 2 7.134470829
1120645 FADS3 -- fatty acid desaturase 3 7.039952979
123
CA 2897828 2019-12-20

Table 2387: MCL vs. PMBL predictor genes
UNIQID Gene name Scale Factor
1132834 SOX11 SRY (sex determining region Y)-box 11 28.17593839
1100873 ESTs 17.90004832
1096503 GL012 -- hypothetical protein GL012 17.43982729
1098840 C3orf6 -- chromosome 3 open reading frame 6 17.37421052
1124734 NA 16.73821457
1135102 PRKCB1 -- protein kinase C, beta 1 16.67436366
1103711 Homo sapiens cDNA FLJ11833 fis, clone HEMBA1006579.
16.57202026
1140416 TOSO -- regulator of Fas-induced apoptosis 15.64802242
1121757 ADRB2 adrenergic, beta-2-, receptor, surface 15.57336633
SPAP1 SH2 domain containing phosphatase anchor
1140236 protein 1 15.20264513
ESTs, Moderately similar to hypothetical protein FLJ20378
1099140 [Homo sapiens] [H.sapiens] 15.11929571
1099549 ESTs 14.92883027
L0058486 transposon-derived Buster1 transposase-like
1139054 protein 14.63422275
1138818 ILF3 interleukin enhancer binding factor 3, 90kDa
14.50621028
ESTs, Highly similar to IL24_HUMAN Interleukin-24
precursor (Suppression of tumorigenicity 16 protein)
(Melanoma differentiation associated protein 7) (M DA-7)
1109444 [H.sapiens] 14.20430672
1124534 K1AA0553 K1AA0553 protein 14.18537487
1098277 PRICKLE1 -- prickle-like 1 (Drosophila) 13.98526258
1131687 TLK1 --tousled-like kinase 1 13.97468703
1125112 PLCL2 phospholipase C-like 2 13.85714318
1125397 Homo sapiens cDNA FLJ33389 fis, clone BRACE2006871.
13.85049805
Table 2388: MCL vs. PTLD predictor genes
UNIQID Gene name Scale Factor
1109603 ESTs 19.95553782
1138222 NA 15.95397369
CD24 CD24 antigen (small cell lung carcinoma cluster 4
1135138 antigen) 15.89198725
RASGRP2 -- RAS guanyl releasing protein 2 (calcium and
1134230 DAG-regulated) 15.80452978
1139411 OSBPL10 oxysterol binding protein-like 10 14.32818885
1140416 TOSO -- regulator of Fas-induced apoptosis 13.89685188
1132834 SOX11 SRY (sex determining region Y)-box 11 13.78424818
1121739 ZNF135 --zinc finger protein 135 (clone pHZ-17) 13.02195529
Human HeLa mRNA isolated as a false positive in a two-
1098156 hybrid-screen. 12.95032505
Homo sapiens cDNA FLJ30555 fis, clone
1099270 BRAWH2003818. 12.7877735
1139012 FLJ20373 -- hypothetical protein FLJ20373 12.70176225
EDG1 -- endothelial differentiation, sphingolipid G-protein-
1120854 coupled receptor, 1 12.25264341
1120985 KIAA0053 KIAA0053 gene product 12.04626201
1115952 L0C146517 --hypothetical protein L0C146517 11.96299478
CHL1 -- cell adhesion molecule with homology to L1CAM
1120825 (close homolog of L1) 11.82402907
1131636 SPOCK2 sparciosteonectin, cwcv and kazal-like 11.80417657
124
CA 2897828 2019-12-20

domains proteoglycan (testican) 2
1136706 MYT1 -- myelin transcription factor 1 11.74962191
1113560 Homo sapiens, clone IMAGE:5725893, mRNA 11.72049882
P4HA1 - procollagen-proline, 2-oxoglutarate 4-
1133851 dioxygenase (proline 4-hydroxylase), alpha polypeptide I -
12.59876059
1137459 BCAT1 - branched chain aminotransferase 1, cytosolic -
14.00465411
Table 2389: MCL vs. SLL predictor genes
UNIQID = Gene name Scale Factor
1132834 SOX11 SRY (sex determining region Y)-box 11 23.59602107
1101987 K1AA1909 KIAA1909 protein 14.50254794
1103711 Homo sapiens cDNA FLJ11833 fis, clone HEMBA1006579.
13.31375894
1096070 DNMT3A -- DNA (cytosine-5-)-methyltransferase 3 alpha
12.37453972
1130926 C5orf13 --chromosome 5 open reading frame 13 11.27840239
1120645 FADS3 --fatty acid desaturase 3 11.14057287
1138099 NA 10.92729287
1097887 KIAA0303 KIAA0303 protein 10.37913127
1099941 ESTs 10.33953409
1130373 KIAA0303 K1AA0303 protein 10.01524528
1110957 SYNE2 spectrin repeat containing, nuclear envelope 2
9.865436185
1130320 ESTs 9.807091644
1124373 LPIN1 lipin 1 9.024985551
1128813 KREMEN2 kringle containing transmembrane protein 2
8.903791941
MARCKS myristoylated alanine-rich protein kinase C
1131130 substrate 8.688979176
CHL1 -- cell adhesion molecule with homology to Li CAM
1120825 (close homolog of L1) 8.685132271
BASP1 - brain abundant, membrane attached signal
1119752 protein 1 8.663402838
1131854 GCLC - glutamate-cysteine ligase, catalytic subunit -
8.761521136
Homo sapiens mRNA; cDNA DKFZp686H1529 (from clone
1105801 DKFZp686H1529) -8.828675125
1097824 MAP2 - microtubule-associated protein 2 -9.345688564
Table 2390: MCL vs. splenic predictor genes
UNIQID Gene name Scale Factor
1106855 K1AA1909 K1AA1909 protein 14.48278638
1121739 ZNF135 - zinc finger protein 135 (clone pHZ-17) 11.95918572
1111850 Homo sapiens cDNA FLJ36977 fis, clone BRACE2006344.
11.13464157
1098024 KIAA1972 KIAA1972 protein 10.10869886
1130764 HNRPAO - heterogeneous nuclear ribonucleoprotein AO
10.06898534
1135342 SHOX2 - short stature homeobox 2 9.565884385
1097218 MGC45400 -- hypothetical protein MGC45400 9.187725705
1117193 RINZF - zinc finger protein RINZF 9.12522795
PSMD10 - proteasome (prosome, macropain) 26S
1139564 subunit, non-ATPase, 10 9.066714773
1132834 SOX11 - SRY (sex determining region Y)-box 11 8.908574745
MARCKS myristoylated alanine-rich protein kinase C
1131130 substrate 8.732921026
PDCD4 - programmed cell death 4 (neoplastic
1131756 transformation inhibitor) 8.441424593
125
CA 2897828 2019-12-20

1102187 DKFZp586C102 ¨ hypothetical protein DKFZp586C1021
8.391861029
1098195 DKFZp762C111 ¨ hypothetical protein DKFZp762C1112
8.349839204
1101211 Homo sapiens cDNA: FLJ21960 fis, clone HEP05517. 8.337208237
1136673 GNAS ¨ GNAS complex locus 8.254076655
1139116 USP16 ¨ ubiquitin specific protease 16 8.179384251
1098694 L0C112868 ¨ hypothetical protein L0C112868 7.935903681
1120519 WWP2 ¨ Nedd-4-like ubiquitin-protein ligase -7.881202253
1114916 FLJ13993 --hypothetical protein FLJ13993 -8.33683119
[00199] With so many candidate predictor genes being utilized, it is possible
to
generate a predictor model that accurately predicts every element of a
training set
but fails to perform on an independent sample. This occurs because the model
incorporates and "learns" the individual characteristics of each sample in the
training
set. Leave-one-out cross-validation was used to verify that the prediction
models
generated above would work on independent samples that the models had not
encountered previously. In this cross-validation method, a single sample is
removed
from the training set, and the predictor is developed again using the
remaining data.
The resulting model is then used to predict the sample that was removed. This
method is repeated with each individual sample taken out. Since no sample is
predicted from a model that includes that sample, this method provides an
unbiased
estimate of predictor accuracy.
[00200] When the predictors developed above were evaluated by leave-one-out
cross-validation, all but one of the 21 MCL samples were correctly identified
as MCL
and none of the 489 non-MCL samples were mistakenly identified as MCL.
Example 13: Identification of lymphoma samples as MCL based on Bayesian
analysis of bene expression data from a Lymphochip microarray:
[00201] Lymphoma samples with morphology consistent with MCL were
identified by pathological review. Since t(11;14) translocation and cyclin D1
overexpression have been consistently associated with MCL, cyclin D1 mRNA
126
CA 2897828 2019-12-20

levels were measured in each sample by quantitative RT-PCR. Of the 101
samples analyzed, 92 expressed cyclin D1 mRNA. These 92 samples, which
were deemed the "core group" of MCLs, were divided into a training set and a
validation set. Gene expression was measured in all 101 samples using a
Lymphochip microarray (Alizadeh 1999). For comparison, gene expression
was measured in 20 samples identified as SLL. In addition, MCL expression
data was compared to expression data obtained previously for GCB (134
cases) and ABC (83 cases) (Rosenwald 2002). Several thousand genes were
differentially expressed between cyclin D1-positive MCL and the other
lymphoma types with high statistical significance (p < 0.001). A complete
listing of these genes is available at-Rosenwald et al., Cancer Cell, 3: 185-
197
(2003), which is referenced therein at page 194 and which is hosted by the
Lymphoma/Leukemia Molecular Profiling Project Gateways at the National
Institute of Health web site.
[00202] Three different binary predictor models were developed: MCL vs. SLL,
MCL
vs. GCB, and MCL vs. ABC. Each of these models was designed to calculate the
probability that a sample was MCL rather than the other lymphoma type in the
pair.
For each pair, the genes that were most differentially expressed between MCL
and
the other lymphoma type in the pair were identified, and the difference in
expression
between the lymphoma types was quantified using a Student's t-test. An LPS was
then calculated for each sample using the following formula:
LPS(X) = trivõ
,E6
127
CA 2897828 2019-12-20

where Xi is the expression of gene j in sample X and tj is the t-statistic for
the
difference in expression of gene j between the two lymphoma types in the pair.
Cyclin D1 was excluded from the calculation of LPS so that the model could be
used
to identify potential MCL cases that were cyclin D1 negative.
[00203] After an LPS had been formulated for each lymphoma sample, the mean
and standard deviation of these LPS's was calculated for each lymphoma type.
For
a new sample X, Bayes' rule can be used to estimate the probability q that the
sample belongs to MCL rather than the second lymphoma type in the pair using
the
following equation:
q(X is MCL) = __________________________________________
cb(LPS(X); /cm, dmci,) + 0(LPS(X); 122 )
where 0(x; ,o-) is the normal density function with mean and standard
deviation o-, and cimc, are the sample mean and variance of the LPS values
for
MCL, and ft, andel-, are the sample mean and variance of the LPS values for
the
second lymphoma type of the pair. A cut-off point of 90% was selected for
assigning
a sample to a particular lymphoma type. Every sample in the training set was
classified correctly using this model (Figure 16). When applied to the
validation set,
the model correctly classified 98% of the cyclin D1-positive MCL cases as MCL
(Figure 16).
[00204] This diagnostic test was applied to nine lymphoma cases that were
morphologically consistent with MCL, but negative for cyclin D1 expression.
Seven
of these samples were classified as MCL, one was classified as GCB, and one
was
not assigned to any lymphoma type because none of the pairs generated a
probability of 90% or greater.
128
CA 2897828 2019-12-20

Example 14: Classification of DLBCL samples based on Bayesian analysis of gene

expression data from the Lymphochip microarrav:
[00205]A statistical method to classify DLBCL samples based on Bayesian
analysis was developed using gene expression data obtained using the
Lymphochip cDNA microarray (Rosenwald 2002). The data was divided into
two sets: a training set used to create and optimize the prediction model, and
a
validation set to evaluate the performance of the model. The training set
consisted of 42 ABC DLBCL samples and 67 GCB DLBCL samples, while the
validation set consisted of 41 ABC DLBCL samples, 67 GCB DLBCL samples,
and 57 type 3 DLBCL samples (Shipp 2002).
[00206] Genes that were listed as present on >50% of the samples were
identified,
and the signal value for these genes on each microarray was normalized to
1,000.
After normalization, all signal values under 50 were set to 50. A 10g2
transformation
was then performed on all the signal values.
(00207] An LPS for distinguishing between two lymphoma types was calculated
for
each sample X in the training set using an equation:
LPS(X) = tiXj,
where Xj represents the expression level of gene] and tj is a scaling factor
whose
value depends on the difference in expression of gene j between the two
lymphoma
types. The scaling factor used in this example was the t-statistic generated
by a t
test of the difference in gene] expression between two lymphoma types. Only
those
genes with the largest t-statistics were included when calculating the LPS for
each
sample. The list of genes used to generate the LPS was narrowed further by
including only those genes that were most variably expressed within the
training set.
129
CA 2897828 2019-12-20

Only genes in the top third with respect to variance were included. Genes that
displayed a correlation with proliferation or lymph node signatures (Shaffer
2001;
Rosenwald 2002) were eliminated from consideration, because these genes are
often variably expressed within samples from a single lymphoma type (Rosenwald
2002).
[00208] Since the LPS is a linear combination of gene expression values, its
distribution within each lymphoma type should be approximately normal,
provided
that it includes a sufficient number of genes and the correlation structure of
those
genes is not extreme. The mean and variance of these normal distributions
within a
lymphoma type can then be estimated from the combined LPS's of all samples
within the type. The LPS distribution of two lymphoma types can be used to
estimate the probability that a new sample belongs to one of the types using
Bayes'
rule. The probability q that a sample Y belongs to lymphoma type 1 can be
determined by an equation:
0(LPS(Y); 11,,ci-,)
q(Y is subtype 1) =
eft(LPS(Y);11d-,)+0(LPS(Y); p2, 2)
where 0(x; , a-) is the normal density function with mean At and standard
deviation
u, lc, and 6 r , are the sample mean and variance of the LPS values for
lymphoma
type 1, and /I, and 62 are the sample mean and variance of the LPS values for
lymphoma type 2. This calculation was used to determine the probability that
each
sample in the training set belonged to GCB or ABC. A sample was classified as
a
particular type if it had a 90% or greater probability of belonging to that
type. The
number of genes in the predictor model was optimized based on the accuracy
with
which the predictor classified samples into the ABC or GCB subtypes defined
previously by hierarchical clustering (Rosenwald 2002). The final predictor
130
CA 2897828 2019-12-20

incorporated 27 genes, and correctly classified 87% of the training set
samples into
the subtype to which they had been assigned by hierarchical clustering (Figure
17).
The genes included in the predictor are listed in Table 2391.
Table 2391
UNIQID Unigene ID Build 167 Gene symbol
19375 235860 FOXP1
19346 109150 SH3BP5
19227 193857 L0C96597
16049 439852 IGHM
32529 55098 C3orf6
24729 127686 IRF4
24899 81170 PIM1
19348 NA NA
27565 444105 ENTPD1
17227 170359 1L16
26919 118722 FUT8
24321 171262 ETV6
29385 167746 BLNK
16858 376071 CCND2
31801 386140 BMF
19234 418004 PTPN1
26385 307734 MME
24361 388737 NA
24570 446198 NA
24904 18166 KIAA0870
24429 155024 BCL6
28224 387222 NEK6
27673 124922 LRMP
24376 317970 SERPINA11
17496 300592 MYBL1
17218 283063 LMO2
28338 78877 ITPKB
[00209] Since the samples used to estimate the distribution of the LPS's were
the
same samples used to generate the model, there was a possibility of
overfitting.
Overfitting would result in a model that indicates a larger separation between
the
LPS's of two lymphoma types than would be found in independent data. To ensure
that overfitting was not taking place, the model was tested on the validation
set. The
reproducibility of the predictor model was verified by its ability to
correctly classify
88% of the samples in the validation set (Figure 18). Interestingly, 56% of
the
DLBCL samples that had been placed in the type 3 subtype by hierarchical
clustering were classified as either ABC or GCB using this Bayesian model.
131
CA 2897828 2019-12-20

[00210] In previous experiments, the genes that were used to distinguish GCB
and
ABC were deliberately selected to include those that were preferentially
expressed in
normal GC B cells (Alizadeh 2000; Rosenwald 2002). In the present analysis,
the
predictor model was not biased a priori to include such genes. The ABC and GCB
lymphoma types as defined by the Bayesian model were analyzed for differential
expression of GC B cell restricted genes. Thirty seven genes were found to be
both
more highly expressed in GC B cells than at other stages of differentiation
(p<0.001)
and differentially expressed between DLBCL subtypes (p<0.001) (Figure 19A).
These 37 genes are listed in Table 2392.
Table 2392
UNIQID Unigene ID Build 167 Gene symbol
28014 300592 MYBL1
24376 317970 SERPINA11
24429 155024 BCL6
16886 124922 LRMP
27374 283063 LMO2
29912 446198
24510 266175 FAG
24854 439767 TOX
32171 307734 MME
24361 388737
19365 171857 Cyorf15a
27292 272251 KLHL5
24822 283794 PCDHGC3
30923 446195
24825 88556 HDAC1
31696 91139 SLC1A1
26976 434281 PTK2
19279 49614 GC ET2
17866 1765 LCK
24386 437459 MY01E
33013 293130 VNN2
25126
30498 157441 SPI1
26512 379414 MFHAS1
26582 153260 SH3KBP1
17840 132311 MAP2K1
26000 25155 NET1
24323 149342 AICDA
30922 435904 C2lorf107
30641 79299 LHFPL2
19308 179608 DHRS9
24455 405387
30034 300208 SEC23IP
24977 169939 HS2ST1
24449 206097 RRAS2
132
CA 2897828 2019-12-20

30763 446198
27987 73792 CR2
All but two (AICDA and DHRS9) of these 37 genes were more highly expressed in
GCB than in ABC. This demonstrates that the DLBCL subtypes defined by the
Bayesian predictor seem to differ with respect to their cell of origin, with
GCB
retaining the gene expression program of normal GC B cells.
[00211]ABC, on the other hand, displayed higher expression of genes
characteristic
of plasma cells (Figure 19B). Twenty four genes were found to be both more
highly
expressed in plasma cells than in B cells at earlier developmental stages
(p<0.001)
and differentially expressed between the DLBCL subtypes (p<0.001). These 24
genes are listed in Table 2393.
Table 2393
UNIQID Unigene ID Build 167 Gene symbol
16614 127686 IRF4
26907 118722 FUT8
31104 313544 NS
19219 355724 CFLAR
26174 28707 SSR3
24566 169948 KC NA3
34500 442808 B4GALT2
26991 314828 UPP1
30191 438695 FKBP11
27402 259855 EEF2K
26096 434937 PPIB
15887 2128 DUSP5
32440 512686 C20orf59
34827 429975 PM5
29232 437638 XBP1
17763 76640 RGC32
32163 445862 RAB30
17814 5353 CASP10
31460 409223 SSR4
26693 83919 GCS1
25130 409563 PACAP
16436 267819 PPP1R2
31610 76901 PDIR
28961 212296 ITGA6
The majority of these plasma cell-restricted genes were more highly expressed
in
ABC than in GCB. Eight of the 32 genes encode proteins that reside and
function in
133
CA 2897828 2019-12-20

the endoplasmic reticulum (ER) or Golgi apparatus, suggesting that ABCs have
increased the intracellular machinery for protein secretion. These eight genes
are
denoted in the above list by the designation "ER" or ''golgi" in parentheses.
Another
gene on this list, XBP-1 transcription factor, encodes a protein that is
required for
plasma cell differentiation (Reimold 2001) and is involved in the response to
unfolded proteins in the ER (Calfon 2002). ABCs have not undergone full
plasmacytic differentiation, however, because other key plasma cell genes such
as
Blimp-1 were not more highly expressed in ABC.
Example 15: Classification of DLBCL samples based on Bayesian analysis of gene

expression data from the Affymetrix HU6800 microarray:
[00212] The prediction method described in Example 14 above was applied to
gene expression data from 58 DLBCL samples obtained using an Affymetrix
HU6800 oligonucleotide microarray (Shipp 2002). The first step in analyzing
this data was to exclude all microarray features with a median signal value of
<200 across the samples. Multiple microarray features representing the same
gene were then averaged. Of the 27 genes in the DLBCL subtype predictor
developed using the Lymphochip data (above), only 14 were represented on
the Affymetrix array and passed this filtering process. These 14 genes are
listed in Table 2394.
Table 2394
UNIQID Unigene ID Build 167 Gene symbol
24729 127686 IRF4
17227 170359 IL16
26907 118722 FUT8
27565 444105 ENTPD1
16858 376071 CCND2
24899 81170 PIM1
16947 418004 PTPN1
16049 439852 IGHM
26385 307734 MME
27673 124922 LRMP
134
CA 2897828 2019-12-20

24429 155024 BCL6
17218 283063 LMO2
28338 78877 ITPKB
17496 300592 MYBL1
These 14 genes were used to create a new DLBCL subtype predictor in which the
LPS scaling coefficients were again calculated based on the DLBCL subtype
distinction in the Lymphochip data set (Rosenwald 2002). To account for
systematic
measuring differences between the Affymetrix and Lymphochip microarrays, the
expression value of each gene on the Affymetrix microarray was shifted and
scaled
to match the mean and variance of the corresponding expression values on the
Lymphochip. The adjusted expression values for each of the 14 genes were then
used to calculate LPS's for each sample. DLBCL subtype membership was again
assigned on a cut-off of 90% certainty. Several observations suggested that
the
predictor identified ABC and GCB samples within the Affymetrix data set that
were
comparable to those found in the Lymphochip data set. First, the relative
proportions of ABC (29%) and GCB (53%) were very similar to the corresponding
proportions in the Lymphochip data set (34% and 49%, respectively). Second, 43
genes were found to be differentially expressed between the two DLBCL subtypes
with high significance (p < 0.001) in the Affymetrix data. This number is
substantially
higher than would be expected by chance, given that the Affymetrix microarray
measures the expression of approximately 5,720 genes. The symbols for these 43
genes were: IGHM; TCF4; IRF4; CCND2; SLA; BATF; KIAA0171; PRKCB1; P2RX5;
GOT2; SPIB; CSNK1E; PIM2; MARCKS; PIM1; TPM2; FUT8; CXCR4; SP140;
BCL2; PTPN1; KIAA0084; HLA-DMB; ACP1; HLA-DQA1; RTVP1; VCL; RPL21;
ITPKB; SLAM; KRT8; DCK; PLEK; SCA1; PSIP2; FAM3C; GPR18; HMG14; CSTB;
SPINK2; LRMP; MYBL1; and LM02. Third, the 43 genes differentially expressed
between the types included 22 genes that were not used in the predictor but
were
135
CA 2897828 2019-12-20

represented on Lymphochip arrays. Fourteen of these 22 genes were
differentially
expressed on the Lymphochip array with high statistical significance (p
<0.001).
Finally, the expression of the c-rel gene was previously found to correspond
to
amplification of the c-rel genomic locus in DLBCL tumor cells, and oncogenic
event
occurring in GCB but not ABC (Rosenwald 2002). In the Affymetrix data set, c-
rel
was differentially expressed between the two subtypes (p = 0.0025), and was
highly
expressed only in a subset of GCB's.
Example 16: Identification of DLBCL samples as PMBL based on Bayesian analysis

of gene expression data from the Lymphochip microarray:
[00213]310 lymphoma biopsy samples identified as DLBCL by a panel of
hematopathologists were divided into a 36 sample training set and a 274
sample validation set, with the validation set consisting of the DLBCL samples
as classified previously in Example 14. All patients from whom the samples
were derived had been treated with anthracycline-containing multiagent
chemotherapy protocols, with some patients additionally receiving radiation
therapy. The training set was profiled for gene expression using Lymphochip
microarrays comprising 15, 133 cDNA elements as described previously
(Alizadeh 2000). This data is available at=the web site companion for
Rosenwald et al., J. Exp. Med., 198: 851-862 (2003), which is referenced
therein at page 852 and which is hosted by Lymphoma/Leukemia Molecular
Profiling Project Gateway at the National Institute of Health web site. The
validation set had previously been profiled using Lymphochip microarrays
comprising 12,196 cDNA elements (Rosenwald 2002). This data is available at
the web site companion for Rosenwald et al., New Eng. J. Med., 346: 1937-
136
CA 2897828 2019-12-20

1947 (2002), which is referenced therein at page 1938 and which is hosted by
the Lymphoma/Leukemia Molecular Profiling Project Gateway at the National
Institute of Health web site.
[00214]A hierarchical clustering algorithm (Eisen 1998) was used to organize
the
genes by their expression patterns across the 36 samples in the training set.
A large
group of genes that were more highly expressed in lymphomas with mediastinal
involvement than in other DLBCLs was shown to be tightly clustered in the
resulting
dendrogram (Figure 20A). This cluster of genes included two genes, MAL and
FIG1,
previously shown to be highly expressed in PMBL (Copie-Bergman 2002; Copie-
Bergman 2003). Several of the lymphomas with mediastinal involvement did not
express this set of putative PMBL signature genes, and it was suspected that
these
samples were more likely to be conventional DLBCL than PMBL. Hierarchical
clustering was used to organize the samples according to their expression of
the
PMBL signature genes, resulting in two major clusters of cases (Figure 20B).
One
cluster contained 21 samples designated "PMBL core" samples by virtue of their
higher expression of PMBL signature genes. The other cluster contained some
samples that had virtually no expression of these genes, and other samples
that did
express these genes but at lower levels than the PMBL core samples.
[00215] A gene expression-based method for distinguishing PMBL core cases from
GCB and ABC DLBCL cases based on Bayesian analysis was developed using the
methods described in Examples 14 and 15. A set of genes were selected that
were
differentially expressed between the PMBL core samples and both GCB and ABC (p
<0.001). This set of genes included all of the PMBL signature genes identified
by
hierarchical clustering (Figure 20A), as well as a large number of additional
genes.
Many of the genes in this set belonged to the lymph node gene expression
signature
137
CA 2897828 2019-12-20

(Alizadeh 2000; Rosenwald 2002). These genes were excluded from the final
predictor because they might cause some DLBCL samples with higher expression
of
lymph node gene expression signature genes to be misclassified as PMBL. The
list
of PMBL distinction genes was refined by adding a requirement that they also
be
differentially expressed between the PMBL core samples and a subgroup of six
DLBCL samples with higher expression of lymph node gene expression signature
genes (p < 0.001). The resulting set of 46 genes included 35 genes that were
more
highly expressed in PMBL and 11 genes that were more highly expressed in DLBCL
(Figure 21A). The 46 genes in this set were PDL2, SNFT, IL13RA1, FGFR1,
FLJ10420, CCL17/TARC, TNFRSF8/CD30, E2F2, MAL, TNFSF4/0X40 ligand,
IL411/Fig1, IMAGE:686580, BST2, FLJ31131, FCER2/CD23, SAMSN1, JAK2,
FLJ00066, MST1R, TRAF1, SLAM, LY75, TNFRSF6/Fas, FNBP1, TLR7,
TNFRSF17/BCMA, CDKN1A/p21CIP1, RGS9, IMAGE:1340506, NFKB2, KIAA0339,
ITGAM, IL23A, SPIN12, MEF2A, PFDN5, ZNF141, IMAGE:4154313,
IMAGE:825382, DLEU1, ITGAE, SH3BP5, BANK, TCL1A, PRKAR1B, and CARD11.
A series of linear predictor scores were generated based on the expression of
this
gene set. Based on the distribution of linear predictor scores within a
particular
lymphoma type, Bayes rule can be used to estimate the probability that a
particular
sample belongs to either of the two types. An arbitrary probability cut-off of
90% or
greater was used to classify a sample as a particular lymphoma type. All of
the
PMBL core samples were classified as PMBL using this method, as were six of
the
other lymphoma samples with mediastinal involvement. However, nine of the
lymphoma samples with mediastinal involvement were classified as a DLBCL, as
were all of the GCB and ABC samples.
138
CA 2897828 2019-12-20

[00216] In the validation set, 11 samples were identified on clinical grounds
as being
consistent with a diagnosis of PMBL, and the Bayesian model classified nine of
these as PMBL (Figure 21B). Interestingly, 12 of the remaining 263 DLBCL
samples
were classified as PMBL by the predictor. Figure 21B shows that these cases
were
indistinguishable by gene expression from the nine cases diagnosed as PMBL on
clinical grounds. As expected, the average expression of the PMBL predictor
genes
in the 249 samples classified as DLBCL was notably lower than in the 22 PMBL
cases. Thus, PMBL represents a third subgroup of DLBCL than can be
distinguished from ABC and GCB by gene expression profiling.
[00217]Table 2395 compares the clinical parameters of patients assigned to the
PMBL, ABC, and GCB subgroups of DLBCL using this prediction method.
Table 2395
ABC GCB PMBL PMBL PMBL P value
DLBCL DLBCL Training set Validation set All cases
Median age 66 61 33 33 33 4.4E-16
Age <35 5% 10% 52% 56% 53% 7.2E-14
Age 35-60 29% 38% 44% 28% 37%
Age >60 66% 52% 4% 17% 9%
Gender = male 59% 53% 44% 50% 47% 0.38
Female <35 2% 3% 32% 39% 35% 1.1E-12
Male <35 2% 7% 20% 17% 19%
Female 35-60 6% 18% 24% 6% 16%
Male 35-60 , 23% 19% 20% 22% 21%
Female >60 33% 25% 0% 6% 2%
Male >60 34% 27% 4% 11% 7%
PMBL patients were significantly younger than other DLBCL patients, with a
median
age at diagnosis of 33 years compared with a median age of 66 and 61 years for
ABC and GCB patients, respectively. Although there was no significant
difference in
gender distribution among the DLBCL subgroups, young women (<35 years)
accounted for 35% of PMBL patients, more than any other DLBCL subgroup. Young
men (<35 years) were also more frequently represented in the PMBL subgroup,
accounting for 19% of the patients. Correspondingly, older men and women (age
139
CA 2897828 2019-12-20

>60) were significantly underrepresented in the PMBL subgroup. These clinical
characteristics were observed in both the training set and the validation set
of PMBL
cases, demonstrating that the PMBL predictor reproducibly identified a
clinically
distinct subgroup of DLBCL patients.
[00218] The PMBL subgroup defined by the PMBL predictor had a relatively
favorable overall survival rate after therapy (Figure 22). PMBL patients had a
five-
year survival rate of 64%, superior to the 46% rate seen in DLBCL patients as
a
whole (p = 0.0067). The survival of the PMBL subgroup was significantly better
than
the 30% five-year survival rate of the ABC subgroup (Figure 22; p = 5.8E-5),
but only
marginally better than the 59% five-year survival rate of the GCB subgroup (p
=
0.18).
Example 17: Classification of lymphomas into types based on Bavesian analysis
of
gene expression data from the Lymph Dx microarray:
[00219] Based on the clustering of the Lymph Dx microarray signals for the
DLBCL
samples, a cluster of "proliferation signature" genes and a cluster of "lymph
node
signature" genes were identified. The expression of these genes was averaged
to
form a proliferation signature and a lymph node signature. Each gene
represented
on the Lymph Dx microarray was placed into one of three "gene-list categories"
based on its correlation with the proliferation or lymph node gene signatures.
"Proliferation" genes were defined as those genes for which the correlation
between
their expression and the proliferation signature was greater than 0.35. Lymph
node
genes were defined as those genes for which the correlation between their
expression and the lymph node signature was greater than 0.35. The remaining
genes on the array were classified as standard genes. This classification
resulted in
323 proliferation genes and 375 lymph node genes.
140
CA 2897828 2019-12-20

[00220] Two stages of lymphoma classification were performed using the gene
expression data obtained for the above samples using the Lymph Dx microarray.
The general procedure used to classify the samples is presented in flow chart
form
in Figure 1.
[00221] For the first stage of expression analysis, the samples were divided
into five
types: FL, MCL, SLL, FH, and a class of aggressive lymphomas that included
DLBCL and BL. Samples obtained from subjects with other diagnoses (e.g., MALT,
LPC) were omitted from this analysis. Data from the Lymph Dx microarray was
then
used to compare gene expression in each possible lymphoma type pair (e.g., FH
vs.
FL, MCL vs. SLL, etc.). This resulted in the creation of ten "pair-wise
models" (one
for each possible lymphoma type pair) for predicting whether a sample fell
into a
particular lymphoma type.
[00222] For each lymphoma type pair, the difference in expression between the
two
types for every gene on the microarray was calculated, and a t-statistic was
generated to represent this difference. Within each gene-list category
(proliferation,
lymph node, and standard), individual genes were ordered based on the absolute
value of their t-statistic. Only those genes that displayed a statistically
significant
difference in expression between the two types were included in the model.
Those
genes with largest absolute t-statistics in each gene-list category were then
used to
generate a linear predictor score (LPS) for each sample. For a sample X and a
set
of genes G, the LPS was defined as:
LPS(X) = ,
,EG
where X is the expression of gene] in the sample and tj is the t-statistic
representing
the difference in expression of gene] between the two lymphoma types. This
141
CA 2897828 2019-12-20

formulation of LPS, known as the compound covariate predictor, has previously
been used successfully (Radmacher 2002; Rosenwald 2003a; Wright 2003). Other
ways to formulate an LPS include Fisher linear discriminant analysis (Dudoit
2002),
weighted voting (Golub 1999), linear support vector machines (Ramaswamy 2001),
and nearest shrunken centroids (Tibshirani 2002).
[00223] In order to optimize the number of genes used to generate the LPS, a
series
of LPS's were generated for each sample using between five and 100 genes from
each gene-list category. The optimal number of genes is that number which
generates a maximum t-statistic when comparing the LPS of two samples from
different lymphoma types (Figure 23). This optimization procedure was repeated
for
every gene-list category in every pair-wise model, meaning that 30
optimizations
were performed in all.
[00224] It was recognized that for some pair-wise models, it would be useful
to
calculate LPS's using different combinations of gene-list categories. LPS's
were
calculated for each sample using four different combinations. In the first,
LPS was
calculated using the standard genes only. In the second, LPS's were calculated
for
both the standard and proliferation genes, but not the lymph node genes. In
the
third, LPS's were calculated for both the standard and lymph node genes, but
not
the proliferation genes. In the fourth, LPS's were calculated using all three
gene-list
categories.
[00225] Depending on the number of gene-list categories included, between one
and
three LPS's were calculated for each sample in the pair-wise models. Thus,
each
sample could be thought of as a vector in a space of between one and three
dimensions. Since the LPS's were sums of individual expressions, it was
reasonable to approximate the distributions as normal. Multivariate normal
142
CA 2897828 2019-12-20

distributions are defined by two quantities: a mean vector, which indicates
the
average value of each of the models within a given lymphoma type, and a
covariance matrix, which indicates the magnitude and orientation spread of
points
away from this center. Both of these quantities can be estimated empirically
from
the observed data. Figure 24 shows the Standard and Proliferation LPS's for
the FL
vs. DLBCL/BL pair-wise model. The dotted lines indicate the standard
deviations
from the fitted multivariate normal distributions.
[00226]Once the multidimensional distributions have been estimated, Bayes'
rule
(Bayes 1763) can be used to estimate the probability that a given sample
belongs to
one lymphoma type or another. Bayesian analysis of an LPS has been
successfully
employed in the past to distinguish DLBCL subtypes (Rosenwald 2003a, Wright
2003). For a sample X, the probability q of the sample belonging to a first
lymphoma
type rather than a second lymphoma type can be calculated using the formula:
0(LPS(X);
q = 0(LPS(X);11,,O-,)+1)(LPS(X); /12,62)
where LPS(X) is the linear predictor score for sample X, 0(x; , a) is the
normal
density function with mean p and standard deviation a, and di are the mean and
variance of the LPS's for the first lymphoma type, and 2 andci, are the mean
and
variance of the LPS's for the second lymphoma type. Using this equation, a
single
probability q value can be developed for each sample and for each of the four
LPS
combinations. This q value can then be used to classify a sample as a first
lymphoma type, a second lymphoma type, or unclassified. Samples with the
highest
q values are classified as the first lymphoma type, while samples with the
lowest q
values are classified as the second lymphoma type. Samples with middle range q
143
CA 2897828 2019-12-20

values are deemed unclassified. Classifying the samples in this manner
requires
two cut-off points: a lower cut-off point between the second lymphoma type and
unclassified, and an upper cut-off point between unclassified and the first
lymphoma
type. To develop these cut-off points, samples were ordered by their q values,
and
each possible cut-off point between adjacent samples was considered. To ensure
that the cut-off points were reasonable, the lower cut-off point was
restricted to
between 0.01 and 0.5 and the upper cut-off point was restricted to between 0.5
and
0.99.
[00227] Every cut-off point and model combination was analyzed by the
following
equation:
3.99 * [(% of type 1 misidentified as type 2) + (% of type 2 misidentified as
type 1)] + [(% of type 1 unclassified) + (% of type 2 misidentified)].
Using this equation, the cut-off point would be adjusted to allow an
additional error
only if this adjustment resulted in four or more unclassified samples becoming
correctly classified. The final model and cut-off point for a given pair-wise
analysis
was that which minimized this equation. The equation utilizes percentages
rather
than the actual number of cases in order to account for the different number
of
samples in each class.
[00228] All cut-off points between a given pair of adjacent q-values will
produce the
same division of data. Since cut-off point optimality is defined in terms of
dividing
the data into subtypes, all cut-off points between a pair of borderline cases
will be
equally optimal. In choosing where to place the actual cut-off point values,
values
were chosen that would lead to a larger unclassified region. When the lower
cut-off
point was being defined, a value would be chosen that was 1/5 of the way from
the
smallest borderline case to the largest. When the upper cut-off point was
being
144
CA 2897828 2019-12-20

defined, a value would be chosen that was 4/5 of the way from the smallest
borderline case to the largest. Figure 25 illustrates the q-results of
optimizing the
cut-point for the FL versus DLBCL/BL samples. The optimal lower cut-off point
for
these samples was found at q=0.49, while the optimal upper cut-off point was
found
at q-=0.84. Figure 26 indicates how this choice of cut-off points divided the
space of
LPS's.
(00229] The above procedures resulted in a series of pair-wise models for
comparing every lymphoma type to every other lymphoma type. If there are n
types,
then there will be n-1 pair-wise models for each type. Since there were five
lymphoma types in the stage 1 analysis, each type was involved in 4 pair-wise
models. For instance, there were four different pair-wise models for MCL: MCL
vs.
FH, MCL vs. FL, MCL vs. SLL, and MCL vs. DLBCL/BL. For each sample tested,
each pair-wise model will produce one of three possible results: 1) the sample
belongs to the first lymphoma type of the pair-wise model, 2) the sample
belongs to
the second lymphoma type of the pair-wise model, or 3) the sample is
unclassified.
If each of the n-1 models agrees that the sample belongs to a particular
lymphoma
type, then the sample is designated as belonging to that type. If the n-1
models do
not all agree that the sample belongs to a particular lymphoma type, the
sample is
designated as unclassified.
[00230] To ensure that the above methods did not result in overfitting (i.e.,
models
that fit particular idiosyncrasies of the training set but fail when applied
to
independent data), the models were validated by leave-one-out cross-validation
fashion (Hills 1966). Each sample was removed from the data one at a time, and
a
predictive model was developed as described above using the remaining data.
This
model was then used to predict the sample that was removed. Since the model
145
CA 2897828 2019-12-20

being used to predict a given sample was generated from data that did not
include
that sample, this method provided an unbiased estimate of the accuracy of the
model.
[00231] The results of the leave-one-out predictions are set forth in Tables
2396 and
2397, below. The rows in each table correspond to different sample groups,
while
the columns indicate the prediction results. The standard to which the
prediction
results were compared in this stage was the diagnoses of a panel of eight
expert
hematopathologists who used histological morphology and immunohistochemistry
to
classify the samples. Table 2396 provides classification results for the five
lymphoma types tested (DLBCL/BL, FL, FH, MCL, SLL), while Table 2397 provides
more specific results for classification of subtypes within these five
lymphoma types.
The results set forth in Table 2396 are also summarized in Figure 27.
Table 2396
1:7
1:11
CD F
CO
U) 0
¨I I
c.)
co LL LL ci al
0
DLBCL/BL 249 6 0 0 0 7
262 95% 2% 3%
FL 5 154 0 0 0 14
173 89% 8% 3%
FH 0 0 17 0 0 0 17
100% 0% 0%
MCL 0 0 0 22 0 0 22
100% 0% 0%
SLL 0 0 0 0 14 0 14
100% 0% 0%
146
CA 2897828 2019-12-20

Table 2397
-0
F _
O
-1 (7) 5
-J (..) ea tz
U_ 2 co 12 t)
C e
ABC 78 0 0 0 0 0 78 100% 0% 0%
GCB 77 4 0 0 0 4 85 91% 5% 5%
PMBL 33 0 0 0 0 0 33 100% 0% 0%
Unclassified 27 1 0 0 0 2 30 90% 7% 3%
DLBCL
DLBCL (not yet 14 0 0 0 0 1 15 93% 7% 0%
subclassed)
BL 20 1 0 0 0 0 21 95% 0% 5%
FL grade 1 1 78 0 0 0 3 82 95% 4% 1%
FL grade 2 2 58 0 0 0 3 63 92% 5% 3%
FL grade 3A 2 18 0 0 0 8 28 64% 29% 7%
Combined FL 5 154 0 0 0 14 173 89% 8% 3%
grades 1, 2, 3A
FL grade 3B 2 1 0 0 0 4 7 14% 57% 29%
FL unknown grade 3 11 0 0 0 0 14 79% 0% 21%
FH 0 0 17 0 0 0 17 100% 0% 0%
MCL 0 0 0 22 0 0 22 100% 0% 0%
SLL 0 0 0 0 14 0 14 100% 0% 0%
[00232]As seen in Table 2396, perfect prediction of SLL, MCL, and FH samples
was obtained. The success rate for predicting FL and the aggressive lymphomas
(DLBCL/BL) was also very good, with only 3% of the samples being classified
incorrectly. As seen in Table 2397, perfect prediction was also obtained for
ABC
and PMBL samples within the DLBCL samples.
Example 18: Classification of DLBCL/BL samples into subtypes based on Bayesian

analysis of gene expression data from the Lymph Dx microarrav:
[00233]Samples identified as DLBCL/BL in Example 17 were subdivided into four
types: ABC, GCB, PMBL, and BL. These samples were then used to generate six
pair-wise models using the same procedure described in Example 17. The results
of
the leave-one-out predictions using these pair-wise models are set forth in
Table
147
CA 2897828 2019-12-20

2398, below. These results are also summarized in Figure 28. The rows in the
table
correspond to different sample groups, while the columns indicate the
prediction
results. In this stage, the ability of the prediction method to identify BL
was again
measured against the diagnoses of hematopathologists. The ability of the
prediction
method to identify the various DLBCL subtypes, on the other hand, was measured
against previous studies in which this distinction between subtypes was based
on
gene expression data from a Lymphochip microarray (Alizadeh 2000, Rosenwald
2002, Rosenwald 2003a, Wright 2003).
Table 2398
CD F
ca co en t:
4 2 8 E m g 12 8 11-1
e
ABC 76 0 0 0 2 78 97% 3% 0%
GCB 1 66 2 4 4 77 86% 9% 5%
PMBL 0 2 27 0 4 33 82% 12 % 6%
Unclassified DLBCL 5 9 1 1 11 27 NA 41% 4%
DLBCL (not yet
5 0 1 3 14 NA 21% 7%
subclassed)
BL 0 1 0 18 1 20 90% 5% 5%
FL grade 1 0 1 0 0 0 1
FL grade 2 0 1 0 0 1 2
FL grade 3A 0 2 0 0 0 2
Combined FL grades 1, 2,
0 4 0 0 1 5
3A
FL grade 3B 0 1 0 0 1 2
FL unknown grade 0 1 0 1 1 3
[00234] As seen in Table 2398, only 1 of the 20 BL lymphoma samples was
classified incorrectly. The classification of DLBCL into subtypes was also
quite
effective. All previously identified ABC subtype samples were again assigned
to the
ABC subtype, while only 5% of the GCB samples and 6% of the PMBL samples
were assigned to a different subtype than they were assigned to previously.
148
CA 2897828 2019-12-20

[00235] The above classification was implemented using S+ software and the S+
subtype predictor script contained in the file entitled
"Subtype_Predictortxt," located
in the computer program listing appendix contained on CD number 22 of 22. This
S+ script implements the lymphoma prediction algorithm. When this script is
pasted
into an S+ script window and run in a working directory containing the data
set files
discussed below, it will produce a text file entitled "PredictionResults.txt,"
which
indicates the results of the predictive algorithm. The other files in the
computer
program listing appendix contain the required data sets, in their required
format, for
carrying out the lymphoma type identification described above. The file
entitled
"GeneData.txt" contains the gene expression values for each sample analyzed.
This
file is included in the working directory when the S+ subtype predictor script
is run.
The file entitled "GenelD.txt" contains information about the genes in the
GeneData.txt file, and is also included in the working directory when the S+
subtype
predictor script is run. This file indicates the UNIQID for each gene, as well
as the
extent to which the gene is associated with the lymph node and proliferation
signatures ("LN.cor" and ''pro.cor," respectively). The file entitled
"SamplelD.txt"
contains information about the samples included in the "GeneData.txt" file,
specifically the original classification of all the samples. This file is also
included in
the working directory when the S+ subtype predictor script is run. The file
entitled
"PredictionResults.txt" is an example of the productive output of the
prediction
algorithm.
[00236] After the above model was validated using leave-one-out cross-
validation,
the model was re-fit using all of the data to generate a final predictor that
could be
applied to a new set of data. Tables 2399-2414 indicate for each of the pair
wise
models the list of genes used, the weight given to each of those genes, the
signature
149
CA 2897828 2019-12-20

with which each gene was associated, the mean values and covariance matrices
associated with the subtypes being compared, and the q-value cut-points of the
pair-
wise model.
Table 2399: ABC vs. BL
Signature Scale UNIQID Unigene ID Build 167 Probe set Gene
symbol
Standard -18.87 1101149 517226 229437 at _ BIC
Standard -17.4 1121452 227817 205681 at _ BCL2A1
Standard -16.42 1123163 421342 208991 at
STAT3
Standard -16.2 1121629 41691 205965 at _ BATF
Standard -15 1134095 89555 208018 s at _ _ HCK
Standard -14.75 1132636 306278 204490 s_at
CD44
Standard -14.33 1119939 170087 202825 at AHR
Standard -14.25 1100138 278391 228234_ at TI
RP
Standard -14.02 1128626 501452 219424_at E813
Standard -13.89 1132883 432453 205027 s at MAP3K8
Standard -13.88 1134991 444105 209474 s_at ENTPD1
Standard -13.37 1109913 355724 239625 at
CFLAR
Standard -13.25 1120389 75367 203761 at _
SLA
Standard -12.99 1131497 114931 202295_s_at CTSH
Standard -12.71 1115071 390476 223218 s at MAIL
Standard -12.46 1136329 132739 211675 s_at
HIC
Standard -12.41 1128195 115325 218695 at RAB7L1
Standard -12.37 1124381 440808 212288_at FNBP1
Standard -12.30 1100562 26608 228737 at _
C200rf100
Standard -12.24 1101272 179089 229584 at DKFZp434
Standard -12.18 , 1128536 21126 219279_at DOCK10
Standard -11.64 1098271 300670 226056_at CDGAP
Standard -11.41 1119566 433506 201954_at ARPC1B
Standard -11.11 1120651 80205 204269_at PIM2
Standard -10.89 1098952 62264 226841_at K1AA0937
Standard -10.80 1099939 488173 227983_at MGC7036
Standard -10.67 1134270 352119 208284 x at
GGT1
Standard -10.44 1134145 4750 208091 s_at DKFZP564
Standard -10.39 1123437 73090 20963-6- at
NFKB2
Standard -10.17 1119884 418004 202716 at
PTPM1
Standard -10.14 1129269 62919 220358_at SNFT
Standard -10.13 1126293 504816 215346_at TNFRSF5
Standard -10.12 1112344 163242 242406 at
Standard -10.10 1135550 221811 210550_s_at RASGRF1
Standard -10.08 1135165 170359 209827 s at
IL16
Standard -10.05 1120808 127686 204562 at IRF4
Standard -10.01 1122087 72927 206693_at IL7
Standard -9.97 1132004 415117 203217_s_at SIAT9
Standard -9.88 1114824 193370 222762_x_at LIMD1
Standard -9.87 1132034 410455 203271 s at UNC119
Standard . -9.87 1099680 210387 227677 at JAK3
Standard -9.86 1132830 31210 204908 s_at BCL3
Standard -9.79 1099631 367639 22762-4 at FLJ20032
Standard -9.78 1120267 256278 203508 at TNFRSF1B
Standard -9.77 1124187 378738 211986_at MGC5395
Standard -9.73 1108970 140489 238604_at
Standard -9.71 1136216 512152 , 211528 x_at
HLA-G ,
Standard -9.71 1120993 327 20491-2 at VORA
150
CA 2897828 2019-12-20

Standard -9.68 1100847 97411 229070 at _ C6orf105
Standard -9.64 1123413 418291 209575 at ILI ORB
Standard -9.62 1115704 350268 224569_ s_ at I RF2BP2
Standard -9.58 1108237 126232 237753 _at
Standard -9.55 1121695 511759 206082_at HCP5
Standard -9.48 1101905 170843 230345_at
Standard -9.42 1119243 440165 201171 at _ ATP6VOE
Standard -9.39 1140457 210546 221658 s_at 1L21R
Standard -9.32 1098506 193400 22633-3- at I L6R
Standard -9.31 1139805 414362 220230 s at _ _ CYB5R2
Standard -9.30 1139037 173380 218223 s at _ _ CKIP-1
Standard -9.28 1130533 76507 200706 s_at LITAF
Standard -9.15 1098678 386140 226535 _at BMF
Standard -9.04 1133210 434374 205842 s at _ _ JAK2
Standard 9.05 1116432 409362 229356_x_at K1AA1259
Standard 9.17 1097281 7037 224892 at _ PLDN
Standard 9.17 1140018 438482 220917_s_at PWDMP
Standard 9.30 1119997 367811 202951_at STK38
Standard 9.41 1119817 409194 202561 at TNKS
Standard 9.55 1139842 133523 220367 s at _ _ SAP130
Standard 9.64 1132122 307734 203434 s at MME
20120 Standard 9.77 1119258 88556 _ _
9_at HDAC1
Standard 9.80 1128248 234149 218802 at FLJ20647
Standard 10.38 1101211 287659 229513- _at STRBP
Standard 10.52 1123419 170195 209590_at BMP7
Standard 10.71 1133755 404501 207318 s at CDC2L5
Standard 10.80 1128192 102506 218696_ at El F2AK3
Standard 10.85 1124786 22370 212847 at _ NEXN
Standard 10.92 1130114 445084 221965 at MPHOSPH9
Standard 11.00 1126081 309763 215030_at GRSF1
Standard 11.17 1118736 96731 38340 at HIP1R
Standard 11.26 1124613 296720 21259 at AUTS2
Standard 11.43 1125456 300592 213906_at MYBL1
Standard 11.60 1097177 9691 224761 at GNA13
Standard 12.11 1120400 152207 203787 at _ SSBP2
Standard 12.12 , 1139266 76640 218723 s at RGC32
Standard 12.22 1100770 65578 228976_at
Standard 12.73 1131246 153752 201853 s_at CDC25B
Standard 13.48 1096503 21379 22352-2- at C9orf45
Standard 14.50 1124920 6150 213039_at ARHGEF1
Standard 15.03 1128360 445043 218988 at SLC35E3
Standard 15.24 , 1099444 434489 227407_at FLJ90013
Standard 21.03 1134582 78202 208794 s at SMARCA4
Standard
Mean ABC -4179.76 Cut 1 0.20
Mean BL -1894.68 Cut 2 0.80
Covariance ABC 53707.58
Covariance BL 194887.5
Table 2400: ABC vs. GCB
Signature Scale , UNIQID Unigene ID Build 167 Probe set
Gene symbol
Standard -15.31 1122645 158341 207641_at TNFRSF13B
Standard -14.56 1120651 80205 204269_at PIM2
Standard -14.18 1120808 127686 204562_at IRF4
Standard -13.84 1114824 193370 222762_x_at LIMD1
151
CA 2897828 2019-12-20

Standard -13.44 1136687 59943 212345 s at _
_ CREB3L2
Standard -13.12 1139805 414362 220230 _ s _at
CYB5R2
Standard -12.23 1104552 193857 233483_at L0C96597
Standard -12.19 1097236 235860 224837 at _
FOXP1
Standard -12.06 1121629 41691 205965_at BATF
Standard -11.93 1128195 115325 218699 at _
RAB7L1
Standard -11.72 1111503 502910 241383 at _
KBRAS2
Standard -11.66 1134991 444105 209474_ s_ at
ENTPD1
Standard -11.27 1098678 386140 226530 at _ BMF
Standard -10.9 1131074 76894 201572 x at _ _
DCTD .
Standard -10.82 1135165 170359 209827 s at _ _
IL16
Standard -10.7 1132396 118722 203988 s at _ _
FUT8
Standard -10.54 1131541 310230 202369_ s_ at
TRAM2
Standard -10.47 1105759 171262 235056 at _
ETV6
Standard -10.38 1121564 437783 205865 at _
ARID3A
Standard -10.16 1130472 192374 200599 s at _ _
TRA1
Standard -10.04 1132058 161999 203313 s_at
TGIF
Standard -10.03 1105684 195155 234973-_at SLC38A5
Standard -9.95 1097735 , 26765 225436_at
L0058489
Standard -9.94 1115071 390476 223218_s_at MAIL
Standard -9.85 1101149 517226 229437_at BIC
Standard -9.83 1119884 418004 202716_ at PTPN1
Standard -9.71 1134095 , 89555 208018 s at _ _
HCK
Standard -9.68 1135550 221811 210550 s_at
RASGRF1
Standard -9.61 1098927 356216 226817_ at FLJ20202
Standard -9.6 1120389 75367 203761 _at SLA
Standard -9.58 1133910 167746 207655 s_at
BLNK
Standard 9.56 1118736 96731 38340- at HIP1R
Standard 9.58 1128860 323634 2197531at STAG3
Standard 9.68 1134582 78202 208794 s_at
SMARCA4
Standard 9.7 1121853 98243 20631-0_at SPINK2
Standard 10.14 1119258 88556 201209_at HDAC1
Standard 10.19 1132122 , 307734 203434_ s_ at
MME
Standard 10.23 1120400 152207 203787 at
SSBP2
Standard 10.48 1529344 317970 Lymph_Dx_-065_at SERPINA11
Standard 10.64 1124613 296720 212599_at AUTS2
Standard 10.72 1132159 147868 203521 _ s_ at
ZNF318
Standard 10.98 1097901 266175 225626 at _ PAG
Standard 11.1 1128287 300063 218862 at _ ASB13
Standard 12.26 1099686 117721 227684_at
Standard 12.45 1112674 310320 242794_at MAML3
Standard 13.15 1120370 78877 203723_at ITPKB
Standard 14.23 1125456 300592 213906_at MYBL1
Lymph Node 6.8 1097202 386779 224796 _at DDEF1
Lymph Node 6.85 1131755 241257 202729_ s_ at LTBP1
Lymph Node 7.27 1136273 13775 211597 s_at HOP
Lymph Node 7.35 1119424 75485 201599-_ at OAT
Lymph Node 7.86 1095985 83883 222450_ at TMEPAI
Lymph Node 8.02 1124875 18166 212975 _at K1AA0870
Lymph Node 8.32 1124655 79299 212658 _at LHFPL2
Lymph Node 8.62 1115034 , 387222 223158 _ s _ at NEK6
Proliferation -9.11 1120583 ,., 153768 204133 _at
RNU3IP2
Proliferation -7.87 1135492 _ 408615 210448 s_at P2RX5
Proliferation -7.68 1127756 313544 21785-0_at NS
152
CA 2897828 2019-12-20

Proliferation -7.57 1097195 149931 224785_at MGC29814
Proliferation -7.31 1127813 14317 217962_at NOLA3
Proliferation -7.24 1138944 84753 218051_s_at FLJ12442
Proliferation -6.99 1139226 266514 218633 x at _ _ FLJ11342
Proliferation -6.7 1137486 441069 214442 s at _ _ MIZ1
Proliferation -6.51 1133786 153591 207396 _ s_ at ALG3
Proliferation -6.45 1131150 75514 201695 _ s_ at NP
Proliferation -6.45 1119076 268849 200681 _at GLO1
Proliferation -6.38 1115679 8345 224523_ s _at MGC4308
Proliferation -6.34 , 1110223 212709 239973_at
Proliferation -6.3 1529338 284275 Lymph_Dx_058_s_a PAK2
t
Proliferation -6.24 1135164 458360 209825_ s_ at UMPK
Proliferation -6.24 , 1128738 335550 219581 at _ MGC2776
Proliferation -6.01 1099088 14355 226996 _at
Proliferation -5.98 1123192 315177 209100 _at IFRD2
Proliferation -5.83 1116073 146161 227103 _ s _at MGC2408
Proliferation 5.79 1097388 278839 225024 _at C20or177
Proliferation 6.13 1124563 249441 212533 _at WEE1
Standard Lymph Node Proliferation
Mean ABC -2226.57 476.67 -1096.34 Cut 1
0.50
Mean GCB -1352.02 547.18 -1005.72 Cut 2
0.74
Covariance ABC 33472.10 3418.91 4347.99
3418.91 1296.05 846.32
4347.99 846.32 1609.13
Covariance GCB 53751.59 466.34 751.08
466.34 777.74 249.29
751.08 249.29 1708.67
Table 2401: ABC vs. PMBL
Signature Scale UNIQID Unigene
ID Build 167 Probe set Gene Symbol
Standard -14.61 1097236 235860 224837_at
FOXP1
Standard -14.47 1104552 193857 233483 _at
L0C96597
Standard -13.62 1122645 158341 207641_at
TNFRSF13B
Standard -12.05 1135102 349845 209685_s_at
P R KC B1
Standard -11.65 1096499 293867 223514_at
CARD11
Standard -11.26 1124770 153261 212827_at
IGHM
Standard , -11.25 1125010 , 43728 213170_at
GPX7
Standard -11.13 1109545 63187 239231_at
Standard -10.99 1109220 445977 238880_at ,
GTF3A
Standard -10.87 1131074 76894 201572_x_at DCTD
Standard -10.68 1134517 75807 208690_s_at
PDLI M1
Standard -10.63 1098604 32793 226444_at
SLC39A10
Standard -10.56 1131219 109150 201810_s_at
SH3BP5
Standard -10.52 1120651 80205 204269_at
PIM2
Standard -10.39 1133910 167746 207655_s_at
BLNK
Standard -10.32 1099396 435949 227346_at
ZNFN1A1
Standard -10.25 1529297 132335 Lymph_Dx_01
at
Standard -10.17 1107575 424589 2370-33_at
MGC52498
Standard -10.11 1117211 356509 233955_x_at
HSPC195
Standard 10.06 1129517 -33 220712_at
153
CA 2897828 2019-12-20

Standard 10.29 1139950 437385 220731 s_at
FLJ10420
Standard 10.35 1097553 197071 22521-4_at
PSMB7
Standard 10.41 1119516 6061 201834_at PRKAB1
Standard 10.47 , 1122772 66742 207900_at CCL17
Standard 10.55 1132762 80395 204777 _ s_ at
MAL
Standard 10.77 1099265 375762 227193_ at
Standard 10.81 , 1095996 , 288801 222482_at SSBP3 ,
Standard 11.14 1100770 65578 228976_at
Standard 11.19 1133801 181097 207426 s_at
TNFSF4
Standard 11.61 1099154 97927 227066-_at
MOBKL2C
Standard 11.63 1120370 78877 203723_at
ITPKB
Standard 11.8 1112674 310320 242794_at
MAML3
Standard 12.57 1105178 283961 234284_at
GNG8
Standard 12.63 1124613 296720 212599 at _
AUTS2
Standard 13.28 1106415 169071 235774_at
Standard 13.3 1121762 32970 206181_at
SLAMF1
Standard 13.6 1121853 98243 206310 at
SPINK2
Lymph Node 10.91 1105838 129837 235142 at _
ZBTB8
Lymph Node 10.99 , 1136273 13775 211597 _ s_ at HOP
Lymph Node 11.02 1099418 172792 227370_ at
K1AA1946
Lymph Node 11.46 1124875 18166 212975_ at
KIAA0870
Lymph Node 11.99 1120299 79334 203574_ at
NFIL3
Lymph Node 12.49 1135871 104717 211031 s at
CYLN2
Lymph Node 13.33 1121767 458324 20618-7 - _at
PTGIR
Proliferation -13.17 1138944 84753 218051_s_at
FLJ12442
Proliferation -11.61 1116122 42768 227408 s_at
DKFZp76100113
Proliferation -11.16 1110223 212709 23997-3_at
Proliferation -9.93 1120717 444159 204394_at
SLC43A1
Proliferation -9.54 1110099 116665 239835_at
TA-KRP
Proliferation -9.49 1130942 445977 201338_x_at
GTF3A
Proliferation -9.28 1123192 315177 209100_at
IFRD2
Proliferation -9.14 1135492 408615 210448 s_ at
P2RX5
Proliferation -9.03 1120011 3068 , 20298-3_at ,
SMARCA3
Proliferation -9.01 1096738 87968 223903_at
TLR9
Proliferation -8.91 1108961 292088 238593_at
FLJ22531
Standard Lymph Node Proliferation
Mean ABC -849.47 531.79 -1027.48 Cut 1 0.20
Mean PMBL 27.99 750.84 -872.43 Cut 2 0.80
Covariance ABC 14028.46 3705.84 3118.60
3705.84 2326.91 1083.37
3118.60 1083.37 1589.42
Covariance PMBL 19425.29 5109.98 2199.28
, 5109.98 2084.28 620.86
2199.28 620.86 1028.44
Table 2402: BL vs. GCB
Signature Scale UNIQID Unigene ID Build 167 Probe set Gene Symbol
Standard -12.78 , 1131246 153752 201853 s_at CDC25B
Standard -11.35 1099444 , 434489 22740-7_at
FLJ90013
Standard -10.4 1116432 409362 229356_x_at K1AA1259
Standard -10.3 1134582 78202
208794_s_at SMARCA4
Standard -10.01 1133998 76884 207826 s_at' 1D3
Standard -9.3 1126081 309763 21503-0_at GRSF1
154
CA 2897828 2019-12-20

Standard -9.19 1096503 21379 223522 _at
C9orf45
Standard -8.95 1529340 -99 Lymph_Dx_06
1 at
Standard -8.88 1138128 390428 21619-
9_ s _at MAP3K4
Standard -8.8 1099152 351247 227064_ at MGC15396
Standard -8.69 1133757 6113
207320_x_at STAU
Standard -8.54 1116593 _ 422889
230329_ s_ at NUDT6
Standard -8.4 1130926 508741 201310_ s _at C5or113
Standard -8.39 1135685 371282 210776
_ x_ at TCF3
Standard -8.39 1140520 , 11747 221741
_ s_ at C20orf21
Standard -8.34 1119802 7370 202522_at
PITPNB
Standard -8.31 1096149 410205 222824_ at
NUDT5
Standard -8.23 1124786 22370 212847 at
NEXN
Standard -8.07 1098012 355669 225756-_at CSNK1E
Standard -7.89 1116317 526415 228661 s at
_ _
Standard -7.86 1109195 416155 238853 _at
Standard -7.71 1134880 168799
209265_ s _at METTL3
Standard -7.66 1529298 136707 Lym ph_Dx_01
6 at
Standard -7.55 1128660 . 413071 2194-
71_at C13orf18
Standard -7.55 1138973 11270 218097
s_at C10orf66
Standard -7.46 1127294 421986 21702-8_at
CXC R4
Standard 7.47 1134270 352119 208284_x_at GGT1
Standard 7.48 1120743 79197 204440_ at CD83
Standard 7.5 1098179 163725 225956 at LOC 153222
Standard 7.55 1121400 223474 205599_-at TRAF1
Standard 7.59 1114967 7905 223028 s_at SNX9
Standard 7.6 1122087 72927 20669-3_at I L7
Standard 7.64 1101905 170843 230345_at
Standard 7.77 1120700 410745 204362_at SCAP2
Standard 7.8 1120572 84 204116_at IL2RG
Standard 7.84 1098271 300670 226056_at CDGAP
Standard 7.9 1115073 131315 223220_s_at BAL
Standard 7.9 1133210 434374 205842 s_at JAK2
Standard 8 1129269 62919 22035-8 _at SNFT
Standard 8.01 1131940 1103 203085_ s _at TGFB1
Standard 8.07 1098506 193400 226333 _at IL6R
Standard 8.13 1120601 441129 204166_at K1AA0963
Standard 8.21 1102540 434881 231093_at FCRH3
Standard 8.24 1121695 511759 206082_at - HCP5
Standard 8.33 1136877 409934 212998 x_ at HLA-DQB1
Standard 8.37 1100138 278391 228234- at TIRP
Standard 8.46 1126293 504816 215346 _at TNFRSF5
Standard 8.46 1127805 380627 217947_at CKLFSF6
Standard 8.59 1136573 914 211991 _ s_ at HLA-DPA1
Standard 8.62 1119111 35052 200804_at TEGT
Standard 8.7 1136329 132739 211675 s_at HC
Standard 8.74 1123690 111805 210176-_at TLR1
Standard 8.81 1138677 390440 217436 x_at
Standard 8.89 1113993 131811 24428-6_at
Standard 8.89 1132651 439767 204529 s_at TOX
Standard 8.91 1119566 433506 201954-_at ARPC1B
Standard 9.01 1128626 501452 219424_at EBI3
Standard 9.17 1101272 179089 229584 at DKFZp434H2111
155
CA 2897828 2019-12-20

Standard 9.33 1136777 387679
212671 s_at HLA-DQA1
Standard 9.33 1109756 530304 23945-3_ at
Standard 9.4 1136216 - 512152
211528 _ x_ at HLA-G
Standard 9.4 1124381 440808
212288 _at FNBP1
Standard 9.46 1099680 210387
227677 at _ JAK3
Standard 9.49 1109913 355724
239629_at CFLAR
Standard 9.55 , 1132636 306278
204490_ s_ at CD44
Standard 9.59 1119243 440165
201171_at ATP6VOE
Standard 9.72 1101149 517226
229437_at BIC
Standard 9.8 1130674 381008
200905 x_at HLA-E
Standard 10.34 1119939 170087 20282-0_at AHR
Standard 10.44 1132883 432453
205027_ s _at MAP3K8
Standard 10.74 1121452 227817
205681 at _ BCL2A1
Standard 10.84 1137360 429658
214196 s at _ _ CLN2
Standard 12.08 1132520 283063
204249 s at LMO2
_ _ _
Standard 12.33 1131497 114931 202295_s_at CTSH
Standard 13.58 1123163 421342
208991 _at STAT3
Lymph Node -9.1 1138136 433574
216215 _ s _at RBM9
Lymph Node 8.78 1130121 411958
221978 at _ HLA-F
Lymph Node 9.22 1139830 221851
220330 _ s_ at SAMSN1
. Lymph Node _ 923 1131705 386467 202638 s , _ _at
ICAM1
Lymph Node 9.62 1130168 75626
222061 at _ CD58
Lymph Node 9.66 1121844 83077
206295_at IL18
Lymph Node 9.68 1121000 519033
204924 at _ TLR2
Lymph Node 9.83 _ 1102437 437023 230966
at _ IL411
Lymph Node 10.71 1119475 296323 201739
at _ SGK
Lymph Node 11.09 1131786 375957
202803_ s_ at ITGB2
Proliferation -11.07 , 1133141 ,
344524 205677_s_at DLEU1
Proliferation -10.04 1138259 89525 216484_x_at HDGF
Proliferation -9.74 1131578 202453
202431_s_at MYC
Proliferation -9.45 1137449 223745
214363_ s_ at MATR3
Proliferation -9.43 1130468 166463
200594_x_at HNRPU
Proliferation -9.21 1138157 82563
216251 s_at KIAA0153
Proliferation -9.15 1127756 313544 21785-
0 _at NS
Proliferation -9 1130433 246112
200058 s_at U5-200KD
Proliferation -8.76 1123108 108112 20882-
8_at POLE3
Proliferation -8.75 1128738 335550
219581_at MGC2776
Proliferation -8.74 1122400 439911
207199 at TERT
Proliferation -8.66 1097948 69476
225684 _at L0C348235
Proliferation -8.6 1119460 76122
201696 at _ SFRS4
Proliferation -8.6 1136401 27258 211761
_ s _at SIP .
Proliferation -8.58 1099088 14355 226996 _at
Proliferation -8.51 1134653 253536
208901 _ s _at TOP1
Proliferation , -8.49 1140584 294083
221932 s , _ _at C14orf87
Proliferation -8.43 1121309 23642
205449_ at H5U79266
Proliferation -8.43 1120385 36708
203755_at , BUB1B
Proliferation -8.38 1136710 75782
212429_s_at GTF3C2
Proliferation -8.36 1136605 448398 212064
x_at MAZ
Proliferation -8.24 1120697 , 323462 20435-
5_at DHX30
Proliferation -8.19 1127833 382044
218001_at MRPS2
Proliferation , -8.11 1096903 437460 224185_at FLJ10385
Proliferation -8.1 1120596 4854 204159_at
CDKN2C
Proliferation -8.1 1120779 28853
204510_at CDC7
156
CA 2897828 2019-12-20

Standard Lymph Node Proliferation
Mean BL 1098.69 576.05 -2392.12 Cut 1 0.09
Mean GCB 2187.37 768.53 -2129.35 Cut 2 0.53
Covariance BL 75263.67 12684.43 15734.77
12684.43 2650.81 2358.05
15734.77 2358.05 4653.00
Covariance GCB 50548.22 9301.12 14182.83
9301.12 2602.51 3028.21
14182.83 3028.21 5983.04
Table 2403: BL vs. PMBL
Signature Scale UNIDID Unigene ID Build 167 Probe set Gene
Symbol
Standard -13.54 1099444 434489 227407 _at FLJ90013
Standard -13.42 , 1096503 21379 223522 _at C9orf45
Standard -13.36 1130114 445084 221965_ at
MPHOSPH9
Standard -13.27 1124786 22370 212847 _at NB(N
Standard -13.27 1134582 78202 208794_s_at SMARCA4
Standard -12.37 1096149 410205 222824_ at
NUDT5
Standard -11.95 1130855 77515 201189_s_at
ITPR3
Standard -11.66 1529298 136707 Lymph_Dx_016
at
Standard -11.35 1131246 153752 201853_s_at CDC25B
Standard -11.17 1136925 436939 213154 _ s _at BICD2
Standard -11.08 1124188 282346 211987_at TOP2B
Standard -11.06 1133998 76884 207826 _ s _at ID3
Standard -10.76 1139266 76640 218723_ s_ at RGC32
Standard -10.74 1134880 168799 209265_ s _at METTL3
Standard -10.69 1140520 11747 221741_s_at C200rf21
Standard -10.6 1109545 63187 239231_at
Standard -10.55 1106043 266331 _ 235372_at FREB
Standard -10.52 1110214 144519 239964 _at TCL6
Standard -10.49 1098592 283707 226431_at
ALS2CR13
Standard -10.45 1109220 445977 238880_at GTF3A
_
Standard -10.41 1131263 249955 201877_s_at PPP2R5C
Standard 10.54 1122772 66742 207900_at CCL17
Standard 10.59 1109913 355724 239629_at CFLAR
Standard 10.82 1119884 418004 202716_at PTPN1
Standard 10.83 1135189 137569 209863 s at TP73L
Standard 10.89 1123437 73090 _ _
_ 209636_at NFKB2
Standard 11.15 1124381 440808 212288_at FNBP1
Standard 11.26 1108237 126232 ' 237753_at
Standard 11.34 1101149 517226 229437 at _ SIC
Standard 11.77 1139774 15827 220140_s_at
SNX11
Standard 11.87 1123163 421342 208991_at STAT3
Standard 11.93 , 1129269 62919 220358_at SNFT
Standard 12.03 1132636 306278 204490_s_at
CD44
Standard 12.1 1138677 390440 217436_x_at
Standard 12.2 1139950 , 437385 220731_s_at FLJ10420
Standard 12.25 1134270 352119 208284_x_at
GGT1
Standard 12.27 1136216 512152 211528_x_at HLA-
G
Standard 12.79 1121400 223474 205599_at TRAF1
Standard 12.82 1119939 170087 202820_at AHR
157
CA 2897828 2019-12-20

Standard 13.12 1126293 504816 215346_ at TNFRSF5
Standard 13.44 1100138 278391 228234 _at
TIRP
Standard 13.74 1132883 432453 205027_s_at MAP3K8
Standard 13.94 1131497 114931 202295 s_at
CTSH
Standard 14.15 1121762 32970 20618-1 _at SLAMF1
Standard 14.51 1132520 283063 204249 s_at
LMO2
Standard 14.68 1121452 227817 20568-1 _at
BCL2A1
Standard 15.24 1105178 283961 234284 at _
GNG8
Lymph Node 10.95 1121205 2488 205269 at LCP2
Lymph Node 11.22 1140845 21486 AF FX----
STAT1
HUMISGF3A/M
97935 3_at
Lymph Node 11.45 1131068 118400 201564 s_at
FSCN1
Lymph Node 11.92 1131705 386467 202638_s_at
ICAM1
Lymph Node 12.06 1131038 81328 201502 s_at
NFKBIA
Lymph Node 12.49 1121444 153563 205668-_ at
LY75
Lymph Node 13.01 1123457 446304 209684 _at
RIN2
Lymph Node 13.19 1140404 354740 221584 s_at KCN
MA1
Lymph Node 13.26 1124875 18166 21297-5_ at
KIAA0870
Lymph Node 14.06 1102437 437023 230966 _at
IL411
Lymph Node 14.11 1132766 82359 204781 s_at
TNFRSF6
Lymph Node 15.31 1121767 458324 20618-7 at PTGIR
_
Lymph Node 15.32 1135871 104717 211031_s_at CYLN2
Lymph Node 15.34 1138652 444471 217388_s_at
KYNU
Lymph Node 16.01 1139830 221851 220330_s_at SAMSN1
Standard Lymph Node
Mean BL -66.97 1445.63 Cut 1 0.20
Mean PMBL 1205.38 2041.25 Cut 2 0.80
Covariance BL 35263.67 13424.88
13424.88 7458.56
Covariance PMBL 12064.38 5113.74
5113.74 3216.53
Table 2404: FH vs. DLBCL-BL
Signature Scale UNIQID Unigene ID Build 167 Probe set Gene Symbol
Standard -12.81 1104910 458262 233969_at IGL@
Standard -11.54 1102898 145519 231496 _at FKSG87
Standard -11.46 1117298 449586 234366_x_at
Standard -11.46 1132973 169294 205255_x_at TCF7
Standard -11.22 1133099 88646 205554_s_at DNASE1L3
Standard -10.76 1131531 153647 202350_s_at MATN2
Standard -10.59 1124283 406612 212144_at UNC84B
Standard -10.35 1099847 36723 227867_at L0C129293
Standard -10.22 1136430 102950 211798_x_at IGLJ3
Standard -10.05 1117394 -13 234792_x_at ,
Standard -9.95 1133047 528338 205434 s_at
AAK1
Standard -9.95 1098865 250905 22674-tat , L0051234
Standard -9.82 1108515 98132 238071_at LCN6
Standard -9.8 1131407 154248 202125 s_at ALS2CR3
Standard -9.77 1128469 390817 21917-3_at
FLJ22686
Standard -9.7 1123875 428 210607_at FLT3LG
Standard -9.69 1131875 169172 202965_s_at CAPN6
158
CA 2897828 2019-12-20

Standard -9.69 1135173 3781 209841 s_at LRRN3
Standard -9.48 1099798 411081 22781T _at FGD3
Standard -9.41 1119046 349499 200606 at _ DSP
Standard -9.36 1122449 278694 207277 at _ CD209
Standard -9.34 1114017 133255 244313_ at
Standard -9.34 1122767 652 207892 at _ TNFSF5
Standard -9.24 1123369 79025 209481 _at SNRK
Standard -9.16 1098954 . 128905 226844_ at MOBKL2B
Standard -9.14 1135513 421437 210481 s_ at CD209L
Standard -9.08 1100904 426296 229145 _at L0C119504
Standard -8.99 1122738 , 81743 207840_at CD160
Standard -8.94 1120925 204891 204773_ at 11_11RA
Standard 9.09 1123055 185726 208691 _at TFRC
Standard 9.62 1134858 405954 209226 s_at TNP01
Standard 10.19 1123052 180909 20868-0_ at PRDX1
Standard 10.81 1124178 446579 211969 at _ HSPCA
Lymph Node -10.59 1137597 3903 214721 x_at CDC42EP4
Lymph Node -9.69 _ 1119684 439586 202242-_ at TM4SF2
Lymph Node -9.25 1125593 8910 214180 at MAN1C1
Lymph Node -8.44 1124318 21858 212190_-at SERPINE2
Lymph Node -8.09 1119448 212296 201656_ at I1GA6
Lymph Node -8.07 1125546 125036 214081 _at PLXDC1
Lymph Node -7.7 1097683 132569 225373 _at PP2135
Lymph Node , -7.56 1101305 112742 229623_ at
Lymph Node 7.45 1135240 436852 209955 _ s _at FAR
Proliferation 6.97 1135101 20830 209680_ s_ at KIFC1
Proliferation 7.03 1130426 432607 200039_s_at PSMB2
Proliferation 7.04 1130501 2795 200650_ s_ at LDHA
Proliferation 7.08 1130744 158688 201027_ s _at ElF5B
Proliferation 7.23 1137506 75258 214501_s_at H2AFY
Proliferation , 7.32 1131474 95577 202246_s_at CDK4
Proliferation 7.39 , 1130871 159087 201222 s_at RAD23B
Proliferation 7.42 1119375 381072 201489-_at PPIF
Proliferation 7.47 1136595 , 404814 212038_s_at VDAC1
Proliferation 7.7 1135858 90093 211015_s_at HSPA4
Proliferation 7.78 1130527 184233 200692_s_at HSPA9B
Proliferation 7.78 1130820 151777 201144_s_at ElF2S1
Proliferation 7.83 1115829 433213 225253_s_at METTL2
Proliferation 7.84 1134699 439683 208974 x at KPNB1
20351 Proliferation 7.87 1120274 31584 _ _
7_ at MTX2
Proliferation 7.92 1136786 63788 212694 s_at PCCB
Proliferation 7.95 , 1097172 434886 22475-3_at CDCA5
Proliferation 8.4 1138537 , -12 217140 s_at
Proliferation 8.53 1119488 154672 20176-1_at MTHFD2
Proliferation 8.58 1130799 233952 201114_x_at PSMA7
Proliferation 8.72 1135673 82159 210759_s_at PSMA1
Proliferation 9.4 1114679 16470 222503_s_at FLJ10904
Standard Lymph Node Proliferation
Mean FH -2193.59 -588.21 1571.78 Cut 1 0.50
Mean DLBCL-BL -1448.27 -441.91 1735.00 Cut 2 0.92
Covariance FH 6729.73 1223.99 2541.22
1223.99 405.22 293.72
2541.22 293.72 1797.58
159
CA 2897828 2019-12-20

Covariance DLBCL-BL 17675.23 3642.41 4158.43
3642.41 1379.81 1066.48
4158.43 1066.48 2858.21
Table 2405: FH vs. FL
Signature Scale UNIQID Unigene ID Build
167 Probe set Gene Symbol
Standard -11.23 1117298 449586 234366 x at
_ _
Standard -10.62 1121953 38365 206478 at KIAA0125
Standard -10.6 1104910 458262 233969-_at IGL@
Standard -10.39 1136430 102950 211798_ x_ at IGLJ3
Standard -9.96 1129281 395486 220377_at C14orf110
Standard -9.73 1118835 102336 47069 at ARHGAP8
Standard -9.21 1127807 7236 217950-_at NOSIP
Standard -9.05 _ 1128377 371003 219014 _at PLAC8
Standard -8.85 1101004 , 2969 229265 _at SKI
Standard 9.06 1139411 _ 368238 219073 _ s_ at OSBPL10
Standard 9.07 1120789 154729 204524_ at PDPK1
Standard 9.21 1136464 159428 211833 _ s_ at BAX
Standard 9.29 1125279 445652 213575 at TRA2A
Standard 9.45 1529390 79241 Lymph_D-x_12 BCL2
Oat
Standard 9.52 1132022 173911 20324-7 s_ at ZNF24
Standard 9.57 1139645 134051 219757_s_at C14orf101
Standard 9.64 1137561 67397 214639_s_at HOXA1
Standard 9.66 1114893 314623 222891 s at BCL11A
_ _ _
Standard 10.38 1098095 131059 225852 _at ANKRD17
Standard 10.4 1134858 405954 209226 s_at TNP01
Standard 12.65 1101054 173328 22932-2 _at PPP2R5E
Standard 12.79 1124178 446579 211969_at HSPCA
Standard 13.34 1135489 288178 210438_x_at SSA2
Standard
Mean FH 136.43 Cut 1 0.50
Mean FL 640.38 Cut 2 0.99
Covariance FH , 10719.40
Covariance FL 9373.11
Table 2406: FH vs. MCL
Signature Scale UNIQID Unigene ID Build
167 Probe set Gene Symbol
Standard 13.05 1100258 88442 228377 at KIAA1384
Standard 13.43 1529382 371468 Lymph_D-x_11 CCND1
1 at
Standard 13.54 1106855 455101 2362-55 at KIAA1909
Standard 13.73 1529308 193014 Lymph_D-x_02
7 x at
Standard 14.56 1100873 445884 229-10-3_at
Standard 21.12 1132834 432638 _ 204914_s_at SOX11
Lymph Node -8.44 1130378 234434 44783 s at HEY1
209879_ Lymph Node -7.92 1123552 423077 _ _
at SELPLG
_
Lymph Node -7.7 1131218 76753 201809 s at ENG
22537 Lymph Node -7.4 1097683 132569 _ _
225373 _at PP2135
Lymph Node -7.15 1136273 13775 211597_s_at HOP
Lymph Node 14.16 1134532 371468 208711 _ s_ at CCND1
160
CA 2897828 2019-12-20

Standard Lymph Node
Mean FH 451.68 -282.65 Cut 1 0.20
Mean MCFL 863.16 -156.82 Cut 2 0.80
Covariance FH 1617.92 222.89
222.89 271.65
Covariance MCL 3154.38 917.30
917.30 659.94
Table 2407: FH vs. SLL
Signature = Scale UNIQID Unigene ID Build 167 Probe set Gene
Symbol
Standard -13.14 1120765 343329 204484_at PIK3C2B
Standard -12.9 1097897 266175 225622_at PAG
Standard 12.72 1133195 274243 205805 s at _ _
ROR1
Standard 12.74 1140416 58831 221601 s at _ _
TOSO
Standard 13.53 1131687 369280 202606_ s _at
TLK1
Standard 13.57 1107044 163426 236458 _at
Standard 14.43 1529389 79241 Lymph_Dx_119 BCL2
at
Standard 14.51 1129026 135146 220-007 at _
FLJ13984
Standard 14.77 1136987 21695 213370_ s_ at
SFMBT1
Standard 14.79 1137109 469653 213689 x at RPL5
Standard 15.37 1529308 193014 Lymph_D-x:027
x at
Standard 15.82 1120832 57856 204-6-04_at PFTK1
Standard 17.37 , 1135550 221811 210550 s_at
RASGRF1
Standard 18.98 1122864 434384 20819-5_at TIN
Lymph Node -12.89 1123038 119000 208636 at _ ACTN1
Lymph Node -12.8 1130378 234434 44783 _ s_ at
HEY1
Lymph Node , -11.59 1124875 18166 212975 _at
KIAA0870
Lymph Node -11.47 1103497 50115 232231 _at
Lymph Node -10.31 1099358 93135 227300 _at
Lymph Node -10.27 1121129 285401 205159 _at CSF2RB
Lymph Node , -10.23 , 1100249 , 388674 228367 _at HAK
Lymph Node -10.05 1132345 109225 203868 _ s_ at
VCAM1
Lymph Node -9.93 1123401 50130 209550 _at NDN
Lymph Node -9.75 1120500 82568 203979 _at CYP27A1
Lymph Node -9.57 1124318 21858 212190 _at
SERPINE2
Lymph Node -9.48 1120288 17483 203547_ at CD4
Lymph Node -9.45 1123372 195825 209487_ at RBPMS
Lymph Node -9.39 1123376 37682 209496_ at RARRES2
Lymph Node -9.29 1123213 12956 209154 at _ TIP-1
Lymph Node -9.23 1098412 409515 226225_ at MCC
Lymph Node -9.23 1125593 8910 214180_ at MAN1C1
Lymph Node -9.17 1131786 375957 202803_ s _at
ITGB2
Lymph Node -9.04 1097683 132569 225373_ at PP2135
Lymph Node -8.91 1097255 380144 224861 _at
Lymph Node -8.76 1131068 118400 201564 _ s _at
FSCN1
Lymph Node -8.7 1119074 54457 200675_ at CD81
Lymph Node -8.68 1125130 35861 213338 _at RIS1
Lymph Node -8.59 1139661 416456 219806_ s _at
FN5
161
'
CA 2897828 2019-12-20

Standard Lymph Node
Mean FH 1144.02 -2223.71 _ Cut 1 0.20
Mean SLL 1592.27 -1798.11 Cut 2 0.80
Covariance FH 902.56 442.69
442.69 809.90
Covariance SLL 2426.26 2938.58
2938.58 9435.72
Table 2408: FL vs. DLBCL-BL
Signature Scale UNIQID Unigene ID Build 167 Probe set Gene Symbol
Standard -23.03 1124833 356416 212914 at _ CBX7
Standard -22.25 1099204 193784 227121 _at
Standard -22.2 1119766 93231 202423 at _ MYST3
Standard -22.04 1099798 411081 227811 at _ FGD3
Standard -22.01 1102898 145519 231496_at FKSG87
Standard -21.79 1131197 269902 201778_s_at K1AA0494
Standard -21.69 1098415 130900 226230_at K1AA1387
Standard -21.57 1120834 57907 204606_at CCL21
Standard -21.39 1130155 436657 222043_at CLU
Standard -20.98 1100904 426296 229145_ at L0C119504
Standard -20.8 1131531 153647 202350_ s _at MATN2
Standard -20.72 1137582 433732 214683_ s _at CLK1
Standard -20.66 1119782 155418 202478 _at TR B2
Standard -20.59 1122767 652 207892_ at TN FSF5
Standard -20.58 1125001 16193 213158_ at
Standard -20.56 1134921 413513 209341 _ s _at IKBKB
Standard -20.56 1132973 169294 205255 _ x_ at TCF7
Standard -20.53 1136984 498154 213364_ s_ at SNX1
Standard -20.41 1115888 35096 225629 s at ZBTB4
20328 Standard -20.37 1120160 436976 _ _
8_ at KIAA0355
Standard -20.36 1139054 25726 , 218263 s_at
L0058486
Standard -20.31 1130030 301872 221834-_at LONP
Standard -20.08 1133024 436987 205383_s_at ZNF288
Standard -20.05 1124666 526394 212672 at ATM
Standard -19.3 1529397 406557 Lym ph_D-x_12 CLK4
7 s at
Standard -19.16 1116056 243678 2269-13- s_at SOX8
Standard -19.14 1098433 202577 22625-0_ at
Standard -19.1 1123635 408614 210073 at _ SIAT8A
Standard -18.95 1138920 24395 218002 _ s _at
CXCL14
Standard -18.84 1133099 88646 205554 s_at DNASE1L3
Standard -18.83 1098495 443668 226318-_ at TERG1
Standard -18.64 1100879 119983 229111_at MASP2
Standard -18.59 1120695 385685 204352_at TRAF5
Standard -18.55 1119983 409783 202920_at ANK2
Standard -18.5 , 1101276 1098 229588_at ERdj5
Standard , -18.47 1099140 , 500350 227052 at
Standard -18.46 1529331 374126 Lym p h_D-x_05
1 s at
Standard -18.45 1131752 170133 2027-24- s_at FOX01A
Standard -18.45 1099265 375762 227193-_at
Standard -18.32 1098179 163725 225956_at LOC 153222
Standard -18.29 1119568 269777 201957_at PPP1R12B
162
CA 2897828 2019-12-20

Standard -18.19 1099900 , 444508 227934_ at
Standard -18.17 1119361 391858 201448 _at TIA1
Standard -18.02 1121650 421137 206002 _at GPR64
Standard -17.91 1100911 320147 229152 at C4orf7
Standard -17.86 1529285 348929 Lymph_D-x_00 KIAA1219
2 at
Standard -17.47 1529357 444651 Lymph1Dx_08
1 at
Standard -17.42 , 1131863 2316 20293-6 s_at SOX9
Standard -17.16 1129943 512828 22162-6_at ZNF506
Standard -17.12 1121301 449971 205437_at ZNF134
Standard -17.11 1131340 437457 202018 s_at LTF
Standard -17.1 1124606 444324 212588-_at PTPRC
Standard -17.08 1131407 154248 202125 _ s_ at ALS2CR3
Standard -16.97 1118939 198161 60528 _at PLA2G4B
Standard -16.91 1134738 , 75842 209033 _ s _at DYRK1A
Standard -16.9 1134083 285091 207996_ s_ at C18orf1
Standard -16.89 1120925 204891 204773_ at ILI 1RA
Standard -16.86 1110070 -101 239803_ at
Standard -16.83 1100042 . 351413 228113 at RAB37
Standard -16.82 1120134 75545 203233_-at IL4R
Standard -16.75 1124283 , 406612 212144_ at UNC84B
Standard -16.72 1109603 -100 239292 _at
Standard -16.71 1120509 155090 204000_at GNB5
Standard -16.65 1133538 , 1416 206760 s_at FCER2
Standard -16.64 1130735 179526 201009-s_at TXNIP
Standard -16.59 1100150 9343 22824-8_ at MGC39830
Standard -16.54 1124237 258855 212080 _at MLL
Standard -16.51 1124416 283604 212331_at . RBL2
Standard -16.48 1133091 73792 205544 _ s _at CR2
Standard -16.46 1131263 249955 201877 _ s _at PPP2R5C
Standard -16.44 1118347 528404 243366_ s _at ITGA4
Standard -16.43 1529343 521948 Lymph_Dx_06
4 at
Standard -16.43 1099549 446665 2275-33_at ,
Standard 17.05 1529453 372679 Lymph_Dx_08 FCGR3A
at
Standard 17.41 1097540 388087 2251-95_ at
Standard 18.47 1140473 , 17377 221676 s_at CORO1C
Standard 18.55 1121100 301921 205098-_at CCR1
Standard 20.07 1124254 301743 212110_at SLC39A14 ,
Standard , 20.2 1130771 61153 201068_ s _at PSMC2
Standard 21.46 1137583 273415 214687 x_ at ALDOA
Standard 21.55 1098168 22151 22594-3_at NLN
Standard 24.07 1123055 185726 208691 _at TFRC
Standard 24.09 1123052 180909 208680 at PRDX1
Lymph Node -20.5 1137597 3903 214721_x_at CDC42EP4
Lymph Node -18.52 1124318 21858 212190_ at SERPINE2
Lymph Node -18.5 1136762 380138 212624_ s_ at CHN1
Lymph Node -18.07 1101305 112742 229623 at
Lymph Node -17.75 1100249 388674 228367_ at HAK
Lymph Node -16.1 1098412 409515 226225 _at MCC
Lymph Node -15.51 , 1140464 111676 221667_ s _at HSPB8
Lymph Node -15.43 1136832 434959 212842_x_at RANBP2L1
163
CA 2897828 2019-12-20

Lymph Node -15.37 1119684 439586 202242_at TM4SF2
Lymph Node -15.02 1097448 250607 225093_ at UTRN
Lymph Node -14.83 1136844 16007 212875_s_at C21orf25
Lymph Node -14.73 1135056 169946 209604 s_at GATA3
Lymph Node -14.48 1097202 386779 22479-6_at DDEF1
Lymph Node -14.44 1121278 21355 205399 _at DCAMKL1
Lymph Node -14.22 1125009 27621 213169_ at
Lymph Node -13.97 1100288 26981 228411 at ALS2CR19
Lymph Node -13.51 , 1132462 14845 204131 _ s_ at
FOX03A
Lymph Node -13.37 1135322 450230 210095_ s _at IGFBP3
Lymph Node -13.35 1097280 423523 224891_at
Lymph Node -12.86 1137097 20107 213656_ s_ at KNS2
Lymph Node -12.85 , 1098809 359394 226682 _at
Lymph Node -12.28 1124875 18166 212975 _at K1AA0870
Lymph Node -12.18 1132345 109225 203868 _ s_ at VCAM1
Lymph Node -12 1097561 19221 225224_ at DKFZP566G142
4
Lymph Node -11.71 1123401 50130 209550 _at NDN
Lymph Node -11.04 1136996 283749 213397 x_ at RNASE4
Lymph Node -10.77 1136788 355455 212698 _ s_ at 36778
Lymph Node -10.71 1098822 443452 226695_ at PRRX1
Lymph Node -10.63 1134200 90786 208161 _ s _at ABCC3
Lymph Node -10.47 1136427 276506 211795 _ s_ at FYB
Lymph Node -10.46 1121186 100431 205242 _at CXCL13
Lymph Node -10.39 1099332 32433 227272 _at
Lymph Node -10.39 1098978 124863 226869_ at
Lymph Node -10.22 1103303 49605 232000 _at C9orf52
Lymph Node , -10.16 1131325 13313 201990 s_at CREBL2
Lymph Node , -10.16 1098174 274401 22594-9_ at L0C340371
Lymph Node -9.93 , 1124733 66762 212771 _at
L0C221061
Lymph Node -9.42 1123372 195825 209487 _at RBPMS
Lymph Node -9.36 1132220 448805 203632 _ s_ at GPRC5B
Lymph Node -9.29 1120703 83974 204368 _at SLCO2A1
Lymph Node -9.26 1132013 434961 203232 _ s _at SCA1
Lymph Node -9.25 1097307 379754 224929_at L0C340061
Lymph Node -9.18 1119251 433941 201194_ at SEPW1
Lymph Node -9.08 1097609 6093 225283 _at ARRDC4
Lymph Node -9.07 1136459 252550 211828_ s _at KIAA0551
Lymph Node -8.86 1132775 1027 204803 s_at RRAD
Lymph Node -8.78 1098946 135121 226834-_ at ASAM
Lymph Node -8.68 1140589 433488 221942_ s_ at GUCY1A3
Lymph Node -8.44 1116966 301124 232744_x_at
Lymph Node -8.39 1100130 76494 228224 _at PRELP
Lymph Node -8.36 1110019 -94 239744_at
Lymph Node -8.3 - 1134647 298654 208892 _ s _at
DUSP6
Lymph Node -8.28 1125593 8910 214180_ at MAN1C1
Lymph Node 7.97 1134370 1422 208438 _ s _at FGR
Lymph Node 8.05 1123566 155935 209906_at C3AR1
Lymph Node 8.09 1131119 349656 201647 s _ at SCARB2
Lymph Node , 8.11 1123586 93841 20994-8 at KCNMB1
Lymph Node 8.13 1128615 104800 219410_ at FLJ10134
Lymph Node , 8.21 1097297 166254 224917 _at VMP1
Lymph Node 8.23 1120299 79334 203574 _at NFIL3
Lymph Node 8.37 1128157 23918 218631 _at VIP32
164
CA 2897828 2019-12-20

Lymph Node 8.4 1130054 82547 221872 _at RARRES1
Lymph Node 8.41 1098152 377588 225922 _at K1AA1450
Lymph Node 8.53 1101566 98558 229947_at
Lymph Node 8.59 1135251 21486 209969 s_at STAT1
Lymph Node 8.84 1099167 381105 227080-_ a= t MGC45731
Lymph Node 9.01 1132920 753 205119 s_at FPR1
Lymph Node 9.26 1097253 77873 224859-_a= t 67H3
Lymph Node 9.29 1120500 82568 203979_ at CYP27A1
Lymph Node 9.36 _ 1131507 172928 202311 s_at
COL1A1
Lymph Node 9.38 1096456 82407 223454-_ a= t CXCL16
Lymph Node 9.49 1136172 38084 211470_s_at SULT1C1
Lymph Node 10.03 1138244 418138 216442_x_at FN1
Lymph Node 10.34 1134424 -17 208540_x_at S100A14
Lymph Node 10.48 1136152 458436 211434 s_at CCRL2
Lymph Node 10.51 1118708 7835 37408-_ at MRC2
Lymph Node 10.6 1136540 179657 211924 s_at PLAUR
Lymph Node 10.63 1098278 166017 22606-6_ at MITE
Lymph Node 10.76 1119477 163867 201743_ at CD14
Lymph Node 10.81 1096429 64896 223405 _at NPL
Lymph Node 11.58 1123672 67846 210152 _at LILRB4
Lymph Node 12 1096364 29444 223276_ at N1D67
Lymph Node 12.16 1119070 445570 200663 _at CD63
Lymph Node 12.3 1133065 77274 205479_s_at PLAU
Lymph Node 12.5 1135240 436852 209955_ s _at FAP
Lymph Node 13.09 1116826 26204 231823 _ s_ at KIAA1295
Lymph Node 13.32 1119068 417004 200660_ at S100A11
Lymph Node 13.45 1120266 246381 203507_at CD68
Lymph Node 13.63 1133216 502577 205872_x_at PDE4DIP
Lymph Node 13.67 1131815 386678 202856_ s_ at SLC16A3
Lymph Node 14.38 1132132 279910 203454 _ s _at ATOX1
Lymph Node 15.25 1134682 411701 208949 _ s_ at LGALS3
Lymph Node 15.46 1119237 389964 201141 _at GPNMB
Lymph Node 15.89 1137698 442669 215001 s at _ _ GLUL
Lymph Node 17.8 1137782 384944 215223 s_ at SOD2
Lymph Node 20.11 1130629 135226 200839 s_at CTSB
Proliferation , 21.02 1119375 381072 20148-9_at PPIF
Proliferation 21.24 1119488 154672 201761 _at MTHFD2
Proliferation 21.31 1119467 21635 201714_at TUBG1
Proliferation 21.68 1130820 151777 201144_s_at ElF2S1
Proliferation 21.69 1131474 95577 202246 s_at CDK4
Proliferation 22.2 1125249 244723 21352-3 _at CCNE1
Proliferation 22.97 1130501 2795 200650_s_at LDHA
Proliferation 23.12 1136913 99962 213113_ s _at SLC43A3
Proliferation 24.05 1130426 432607 200039_s_at PSMB2
Standard , Lymph Node Proliferation
Mean FL -11121.51 -1603.39 1890.60 Cut 1
0.34
Mean DLBCL-BL -8760.65 -460.71 2101.10 Cut 2
0.94
Covariance FL 246359.77 111505.42 28908.20
111505.42 67036.17 13130.59
28908.20 13130.59 4617.24
Covariance DLBCL-BL 413069.12 178811.32 30151.89
178811.32 106324.53 10877.26
30151.89 10877.26 5180.68
165
CA 2897828 2019-12-20

. Table 2409: FL vs. MCL
Signature Scale UNIQID Unigene ID Build 167 Probe set
Gene Symbol
Standard -24.56 1123731 17165 210258_ at RGS13
Standard -22.56 1133192 24024 205801 _ s _ at RASGRP3
,
Standard -21.12 1114543 156189 244887_at
Standard -18.49 1120090 155024 203140_at BCL6
Standard -18.07 1124646 436432 212646 _at RAFTLIN
Standard -17.24 1132122 307734 203434 s_at MME
Standard -16.63 1105986 49614 235310-_ at GCET2
Standard -15.09 1120134 75545 203233_ at 1L4R
Standard -14.05 , 1132651 439767 204529_s_at TOX
Standard 13.8 1098277 6786 226065 _at PRICKLE1
Standard 13.85 1109560 207428 239246_ at FARP1
Standard 13.86 1103504 142517 232239_ at
Standard 13.88 1132734 126248 204724_s_at COL9A3
Standard 13.91 1115905 301478 i 225757 s_at CLMN
Standard 14.89 1098840 55098 22671-3_at C3orf6
Standard 14.97 1100873 445884 229103_ at
Standard 14.99 1139393 170129 219032_x_at OPN3
Standard 16.13 1124864 411317 212960_ at K1AA0882
Standard 16.36 1106855 455101 236255 _at K1AA1909
Standard 16.43 1120858 410683 204647 _at HOMER3
Standard 17.38 1130926 508741 201310 s _ at C5or113
Standard 18.3 1103711 288718 23247-8 at
Standard 18.62 1109505 8162 239186_ at MGC39372
Standard 20.31 1132834 432638 204914_ s _at SOX11
Standard 22.61 1096070 241565 222640_ at DNMT3A
Standard 28.66 1529382 371468 Lymph_Dx_111 CCND1
at
Lymph Node -10.77 1097202 386779 224-
796 _at DDEF1
Lymph Node -10.22 1119546 433898
201921 _at GNG10
Lymph Node -9.89 1132766 82359 204781 _ s
_at TNFRSF6
Lymph Node -9.4 1138867 10706 217892 _ s
_at EPLIN
Lymph Node 9.65 1125025 301094 213196_at
Lymph Node 10.44 1134797 433394 209118 s_at
TUBA3
Lymph Node 22.6 1529456 371468 Lymph_D-x_113
CCND1
at
Proliferation -7.36 1097948 69476 225-6-84_ at
L0C348235
Proliferation -7.31 1130747 234489 201030_ x_ at
LDHB
Proliferation -6.95 1130923 459987 201306 s_at
ANP32B
Proliferation -6.87 1120205 5198 20340-5 _at
DSCR2
Proliferation -6.64 1132468 79353 204147 s_at
TFDP1
Proliferation -6.1 1119916 177584 20278-0_at
OXCT
Proliferation -6.08 1119873 446393 202697_at
CPSF5
Proliferation -6.08 1119488 154672 201761_at
MTHFD2
Proliferation -6.04 1130658 447492 200886_s_at
PGAM1
Proliferation -5.82 1132825 512813 204900_x_at
SAP30
Proliferation -5.53 1115607 435733 224428 _ s_ at
CDCA7
Proliferation -5.44 1120316 63335 203611 _at
TERF2
Proliferation -5.34 1114970 279529 223032_x_at
PX19
Proliferation -5.32 1140843 169476 AFFX- GAPD
HUMGAPDH/M
166
CA 2897828 2019-12-20

33197_5_at .
Proliferation -5.28 1131081 180610 201586 _ s _at SFPQ
Proliferation -5.15 1121062 408658 205034_at CCNE2
Proliferation 5.15 1120986 172052 204886 at PLK4
Proliferation 5.16 1097195 149931 224785-_at MGC29814
Proliferation 5.2 1120011 3068 202983_at SMARCA3
Proliferation 5.47 1100183 180582 228286_at FLJ40869
Proliferation 5.67 1121012 96055 204947_at E2F1
Proliferation 5.84 1115679 8345 224523_s_at MGC4308
Proliferation 5.88 1135285 449501 210024 s_at UBE2E3
Proliferation 5.92 1120520 35120 20402-3_at RFC4
Proliferation 6.16 1529361 388681 Lymph_Dx_086 HDAC3
s at
Proliferation 6.45 1096054 21331 22-26-06_ at FLJ10036
Proliferation 6.45 1096738 87968 223903 at _ TLR9
Proliferation 6.51 1136781 120197 212680_ x_ at PPP1R14B
Proliferation 6.63 1119466 179718 201710_at MYBL2
Proliferation , 6.65 1136285 182490 211615_s_at LRPPRC
Proliferation 6.67 1136853 66170 212922 s_at SMYD2
Proliferation 7.45 1119390 77254 20151-8_at CBX1
Proliferation 8.87 1116122 42768 227408_s_at DKFZp761001
13
Proliferation 10.12 1119515 3352 201833 _at HDAC2
Standard Lymph Node Proliferation ,
Mean FL -18.82 -33.90 23.53 Cut 1 0.14
Mean MCL 1558.10 113.95 165.48 Cut 2 0.58
Covariance FL 21302.14 1098.24 678.04
1098.24 226.29 75.99
678.04 75.99 315.67 '
Covariance MCL 81008.29 5261.37 9185.20
5261.37 2047.34 875.56
9185.20 875.56 1447.43
Table 2410: FL vs. SLL
Signature Scale UNIQID Unigene ID Build 167 Probe set Gene Symbol
Standard -21.04 1123731 17165 210258 at
RGS13
Standard , -20.91 1124646 436432 212646_at
RAFTLIN
Standard -18.82 1099651 120785 227646 at _ EBF
Standard -18.12 1114543 156189 244887 at
- Standard -17.85 1105986 49614 235310_at GCET2
Standard -16.73 1100911 320147 229152_at C4orf7
Standard -15.77 1132122 307734 203434 s_at MME
Standard -15.12 1120090 155024 203140-_at BCL6 ,
Standard -14.89 1097897 266175 225622 at PAG
Standard -14.36 1529343 521948 Lymph_D-x_06
4 at
Standard -14.32 1529318 291954 Lymph-_Dx_03
8 at
Standard -14.06 1128694 171466 2195-17_at ELL3
Standard -13.61 1101586 187884
229971_at GPR114
Standard -13.57 1119752 511745 202391_at BASP1
Standard -13.13 1137561 67397 214639_s_at
HOXA1
167
CA 2897828 2019-12-20

Standard -12.85 1097247 388761 224851 at
CDK6
Standard -12.43 1529344 317970 Lymph_D-x_06 SERPINA11
at
Standard -12.4 1120765 343329 2044-84_ at
PIK3C2B
Standard -12.33 1130155 436657 222043 at CLU
Standard -12.07 1529292 -92 Lymph_D-x_01
0_at
Standard -12.01 1119939 170087 202820_at AHR
Standard -11.82 1119919 199263 202786_at STK39
Standard -11.77 1099686 117721 227684_at
Standard -11.63 1119782 155418 202478 at
TRB2
Standard 10.97 1529309 512797 Lymph_D-x_02 HSH2
8 at
Standard 10.97 1139393 170129 21903-2 _ x_ at
OPN3
Standard 11.04 1131246 153752 201853 _ s_ at
CDC25B
Standard 11.07 1140391 44865 221558_s_at LEF1
Standard 11.16 1140416 58831 221601_s_at TOSO
Standard 11.35 1127807 7236 217950 at NOSIP
Standard 11.67 1529317 -98 Lymph_D-x_03
7_at
Standard 11.81 1117343 306812 234643_x_at
BUCS1
Standard 11.82 1102081 506977 230551 _at
Standard 11.82 1135042 79015 209582 _ s_ at
MOX2
Standard 11.96 1132734 126248 204724_ s_ at
COL9A3
Standard 12.09 1137109 469653 213689_x_at RPL5
Standard 12.14 1099939 , 488173 227983_at MGC7036
Standard 12.19 1129103 99430 220118_at
TZFP
Standard 12.47 1135592 758 210621 _ s _at RASA1
Standard 12.78 1108970 140489 238604 at
Standard 12.92 1097143 74335 224716_at HSPCB
Standard 13.18 1136865 412128 212959 s at
MGC4170
22600 Standard 13.96 1098220 80720 _ _
226002 _at GAB1
Standard 14.06 1100847 97411 229070_at C6orf105
Standard 14.39 1098865 250905 226741 _at L0051234
Standard 15.57 1136687 59943 212345 s_at
CREB3L2
Standard 15.75 1107044 163426 23645-8_at
Standard 16.52 1123622 8578 210051_at EPAC
Standard 17.74 1136987 21695 213370_s_at SFMBT1
Standard 19.15 1129026 135146 220007_at FLJ13984
-
Standard 19.65 1131854 414985 202923 s_at
GCLC
Lymph Node -14.99 1124875 18166 21297-5_ at
KIAA0870
Lymph Node -14.33 1099358 93135 227300 _at
Lymph Node -13.26 1121129 285401 205159_ at CSF2RB
Lymph Node -12.61 1119074 54457 200675 _at
CD81
Lymph Node -12.52 1121029 412999 204971 _at
CSTA
Lymph Node -11.48 1137247 234734 213975_ s _ at
LYZ
Lymph Node -10.97 1128781 79741 219648 _at FLJ10116
Lymph Node 11.79 1119880 442844 202709_at
FMOD
Lymph Node 14.4 1134370 1422 208438 _ s _at FGR
Standard Lymph Node
Mean FL -663.95 -730.08 Cut 1 0.20
Mean SLL 1332.84 -484.93 Cut 2 0.80
168
CA 2897828 2019-12-20

Covariance FL 37097.15 1710.73
1710.73 663.78
Covariance SLL 85989.25 17661.52
17661.52 4555.06
Table 2411: GCB vs. PMBL
Signature Scale UNIQID
Unigene ID Build 167 Probe set Gene Symbol
Standard -8.39 1096440 231320 223423 at _ GPR160
Standard -8.13 1096108 292871 222731_at ZDHHC2
Standard -8.12 1125231 446375 213489 at _ MAPRE2
Standard , -8.02 1136759 188882 212605 s at
_ _
Standard -7.91 1096499 293867 223514 at _ CARD11
Standard -7.8 1099388 124024 227336 at _ DTX1
Standard -7.71 1139623 193736 219667_ s_ at BANK1
Standard -7.68 1098592 283707 226431 _at ALS2CR13
Standard -7.67 1107575 424589 237033_at MG052498
Standard , -7.63 1116829 115467 231840 x_at
L0C90624
Standard -7.42 1130114 445084 22196-5_at MPHOSPH9
Standard -7.27 1098909 446408 226789_at
Standard 7.34 1138759 396404 217707_x_at SMARCA2
Standard 7.37 1120355 80420 203687_at CX3CL1
Standard 7.4 1134270 352119 208284_x_at GGT1
Standard 7.44 1115441 5470 224156 x_at IL17RB
Standard 7.78 1103054 341531 23169-0_at
Standard 7.91 1119765 81234 202421 at _ IGSF3
Standard 7.92 1119438 118110 201641_at BST2
Standard 8.09 1135645 31439 210715 s_at SPINT2
Standard 8.15 1106015 96885 23534-3_ at FLJ12505
Standard 8.18 1121400 223474 205599_at TRAF1
Standard 8.38 1139950 437385 220731 s_at FLJ10420
Standard 8.73 1122112 1314 20672-9_at TNFRSF8
_
Standard 8.77 1122772 66742 207900_at CCL17
Standard 8.84 1132762 80395 204777_s_at MAL
Standard 9.64 1139774 15827 220140_s_at SNX11
Standard 10.53 1133801 181097 207426 s_at TNFSF4
Standard 11.52 1106415 169071 235774-_at
Standard 12.09 1129269 62919 220358_at SN FT
Standard
Mean GCB 292.76 Cut 1 0.16
Mean PMBL 725.28 Cut 2 0.50
Covariance GCB 8538.86
Covariance PMBL 11405.23
169
CA 2897828 2019-12-20

Table 2412: MCL vs. DLBCL-BL
Signature Scale UNIQID Unigene ID Build 167 Probe set Gene Symbol
Standard -26.11 1529382 .. 371468 Lymph_Dx_11 CCND1
1 at
Standard -18.35 1103711 288718 232478_at
Standard -17.03 1106855 455101 236255_at KIAA1909
Standard -16.49 1098840 55098 226713 _at
C3orf6
Standard -15.41 1109505 8162 239186 _at MGC39372
Standard -15.11 1098954 128905 226844 at
MOBKL2B
Standard -14.96 1103504 142517 232239-_at
Standard -14.74 1096070 241565 222640 _at
DNMT3A
Standard -13.81 1137663 247362 214909 _ s_ at
DDAH2
Standard -13.8 1124864 411317 212960 at _
KIAA0882
Standard -13.62 1140127 125300 221044 _ s _at
TRIM34
Standard -13.62 1119361 391858 201448_ at TIA1
Standard -13.37 1127849 76691 218032 at _
SNN
Standard 13.72 1133192 24024 205801_s_at RASGRP3
Standard 13.85 1137583 273415 214687_x_at ALDOA
Standard 15.02 1123052 180909 208680_at PRDX1
Standard 16.21 1097611 438993 225285 at
BCAT1
Lymph Node -19.18 1529456 371468 Lymph_D-x_11
CCND1
3 at
Lymph Node -10.71 1098978 124863 2268-69 _at
Lymph Node -9.17 1097448 250607 225093_at
UTRN
Lymph Node 8.84 1135240 436852 209955 s_at
FAP
Lymph Node 9.11 1119475 296323 20173-9 _at
SGK
Lymph Node 9.22 1119237 389964 201141_at
GPNMB
Lymph Node 9.46 1130629 135226 200839_ s _at
CTSB
Lymph Node 10.1 1130054 82547 221872 _at
RARRES1
Standard Lymph Node ,
Mean MCL -1417.55 -25.58 Cut 1 0.50
Mean DLBCL-BL -756.07 202.29 Cut 2 0.88
Covariance MCL 15347.98 3525.48
3525.48 .. 5420.31
Covariance DLBCL-BL 5132.06 .. 1007.64
1007.64 .. 991.38
Table 2413: MCL vs. SLL
Signature Scale UNIQID Unigene ID Build 167 Probe set Gene Symbol
Standard -20.18 1132834 432638 204914_s_at
SOX11
Standard -15.17 1130926 508741 201310 _ s _at
C5orf13
Standard 13.44 1116150 16229 227606 s_at
AMSH-LP
Standard 14.44 1120134 75545 203233-_at
IL4R
Standard 15.18 1529437 445162 Lymph_Dx_17
BTLA
at
Standard 15.19 1529317 -98 Lymph-_Dx_03
7 at
Standard 16.2 1135042 79015 20958-2_s_at
MOX2
Standard
Mean MCL 181.38 Cut 1 0.20
170
CA 2897828 2019-12-20

=
Mean SLL 564.92 Cut 2 0.80
Covariance MCI 1734.42
Covariance SLL 910.75
Table 2414: SLL vs. DLBCL-BL
Signature Scale UNIQID Unigene ID Build 167
Probe set Gene Symbol
Standard -16.014498 1123622 8578 210051 at EPAC
Standard -15.26356533 1102081 506977 230551_at
Standard -14.82150028 1107044 163426 236458_at
Standard -14,17813266 1098865 250905 226741_at L0051234
Standard -12.92844719 1110740 416810 240538 at
Standard -12.86520757 1129026 135146 220007 at
FLJ13984
Standard -12.2702748 1135592 758 210621
_ s _at RASA1
Standard -11.87309449 1117343 306812 234643_x_at BUCS1
Standard -11.81789137 1136987 21695 213370 s_at
SFMBT1
Standard -11.78631706 1124830 9059 21291-1_at K1AA0962
Standard -11.39454435 1133538 1416 206760_s_at FCER2
Standard -11.39050362 1135802 439343
210944 s_at CAPN3
Standard 11.72928644 1120770 300825 20449-3_at BID
Lymph Node -12.21593247 1119880 442844 202709 at FMOD
Lymph Node 9.514704847 1135240 436852 209955_ s _at FAP
Lymph Node 9.739298877 1096429 64896 223405_at NPL
Lymph Node 10.05087645 1119475 296323 201739_at SGK
Lymph Node 13.11985922, 1119237 389964 201141_at GPNMB
Proliferation 10.47525875 1128106 14559 218542 at
C10orf3
Proliferation 10.53295782 1132825 512813 204900 x at _ _ SAP30
Proliferation 11.93918891 1130501 2795 200650 s_at LDHA
Proliferation 11.98738778 1123439 287472 209642-_at BUB1
Proliferation 11.99741644 1115607 435733 224428_s_at CDCA7
Standard Lymph Node Proliferation
Mean SLL -1383.640809 177.4452398
467.2463569 Cut 1 0.201266305
Mean DLBCL-BL -926.7275468 329.6795845
582.9070266 Cut 2 0.799816116
Covariance SLL 3591.384775 1789.7516 856.0703202
1789.7516 1421.869535 663.4782048
856.0703202 663.4782048 965.6470151
Covariance DLBCL-BL 2922.643347 473.543487 634.3258773
473.543487 931.9845277 -53.85584619
634.3258773 -53.85584619 767.3545404
Example 19: Classification of lymphoma samples as BL or DLBCL based on
Bayesian analysis of gene expression data from the Affvmetrix U133 plus 2.0
microarrav and the Lymphoma microarrav:
[002371Tumor biopsies were obtained from 71 previously untreated, HIV-negative
patients diagnosed with sporadic BL (54 cases) or Burkitt-like lymphoma (17
cases)
between 1986 and 2004 at seven institutions belonging to the Leukemia Lymphoma
171
CA 2897828 2019-12-20

Molecular Profiling Project (LLMPP). In addition, 232 samples were obtained
from
patients diagnosed with DLBCL. 223 of these were part of previously published
studies (Rosenwald 2002; Rosenwald 2003b) using the Lymphochip microarray
(Alizadeh 1999), while the other nine were "high-grade" DLBCL samples with
K167
scores (a measure of lymphoma cell proliferation) of nearly 100%. All 303
cases
were reviewed by a panel of eight hematopathologists using current WHO
criteria for
morphology, immunophenotype, and cytogenetics. Specifically, specimens
classified as BL were required to have a c-myc translocation, morphology
consistent
with BL, a Ki67+ proliferative fraction of greater than 90%, and
immunohistochemical
evidence of CD10 and/or BCL6 expression. Specimens were classified as DLBCL
based on morphological criteria and a B cell immunophenotype.
[00238] Among the 232 DLBCL cases, the median age at diagnosis was 61.5 years
(range, 8.9 to 92 years). Median follow up was 2.5 years (6.8 years for
survivors).
FISH for c-myc translocation was performed in 87 cases, with six cases testing
positive.
[00239] Among the 71 BL cases, 48% were pediatric (range, 2.9 to 18 years) and
the remainder were adults (range, 18 to 73 years). Median follow-up was 1.6
years
(4.9 years for survivors). The regimens used to treat BL were grouped into
CHOP-
like regimens (CHOP (Fisher 1993), CNOP (Vose 2002)) or intensive regimens
(BFM (Pees 1992), CODOX-M IVAC (Magrath 1996), and intensive chemotherapy
regimens combined with autologous stem cell transplant (ASCT)). FISH for c-myc
translocation was performed in 67 of the 71 cases, including all cases in
which BL
was not ruled out by immunohistochemistry or morphology, and 52 cases were
found to be positive. FISH for BCL2 translocation was performed in 44 of the
71
cases, with seven cases testing positive.
172
CA 2897828 2019-12-20

[00240]After pathological review and reclassification, the 71 cases originally
submitted as BL or Burkitt-like lymphoma were divided into classic BL (25
cases),
atypical BL (20 cases), DLBCL (20 cases), and high grade lymphomas that could
not
be classified by current WHO criteria (six cases). This re-review diagnosis,
which is
summarized in Table 2419, provided the standard against which gene expression-
based predictors were measured.
Table 2419:
Submitting
diagnosis Pathological diagnosis Total cases Gene
expression diagnosis Total cases
Classic BL 25 BL 25
BL or Burkitt-like BL 19
lymphoma (71
Atypical BL 20 DLBCL 1
cases)
BL 7
DLBCL 20 DLBCL 13
DLBCL 5
High grade lymphoma (NOS) 6 BL 1
DLBCL (223 cases) ABC 78
GCB 82
PMBL 33
DLBCL 223 Unclassified DLBCL 30
High grade DLBCL ABC 6
(9 cases) GCB 2
DLBCL 9 BL 1
[00241] RNA was extracted from each of the 303 samples as described
previously (Alizadeh 2000). Gene expression profiling was performed using
the custom oligonucleotide Lymphoma microarray, which contains
oligonucleotides corresponding to 2,524 unique genes that are differentially
expressed among the various forms of non- Hodgkin's lymphoma. A subset of
the samples were profiled on whole-genome Affymetrix U133 plus 2.0 arrays
as well.
(00242] To develop a gene expression-based diagnosis of BL, the initial focus
was
on those cases that were submitted as BL and confirmed as such by pathological
173
CA 2897828 2019-12-20

review (45 cases). A set of genes were identified that were differentially
expressed
between these BL cases and each of the DLBCL subgroups (Figure 35A). Pair-wise
Bayesian compound covariate predictors were constructed between BL and ABC,
BL and GCB, and BL and PMBL as described previously (Rosenwald 2003a;
Rosenwald 2003b; Wright 2003). For each comparison of two lymphoma types, a
linear predictor score was calculated by:
LPS(S) = tiSj,
where Si is the expression of gene j in a sample S and ti is a scale factor
representing the difference in expression of gene j between a first lymphoma
type
and a second lymphoma type (Radmacher 2002). The scale factor used was the t-
statistic generated by a t-test for the difference in expression between the
two
subtypes being compared. Bayes' rule was then applied to the distribution of
the
linear predictor scores to estimate the probability that the sample was a
member of
either group. Each comparison between BL and a DLBCL subtype proceeded in two
stages using different sets of genes to create a compound covariate predictor
as
described above.
[00243] Stage one utilized c-myc and its target genes, which were defined
using an
RNA interference experiment. For the RNA interference experiment, the OCI-Ly10
DLBCL cell line was transfected by electroporation (Amaxa Inc., Gaithersburg,
MD)
with small interfering RNAs (siRNAs) targeting the c-myc gene (Smart pool;
Dharmacon, Lafayette, CO). Gene expression in RNAi-transduced OCI-Ly10 cells
was compared to that of control-transfected OCI-Ly10 cells using Lymphochip
microarrays (Alizadeh 1999). Genes that were at least 40% downregulated at 16
and 18 hours post-RNAi transfection and whose mRNA expression levels were
174
CA 2897828 2019-12-20

correlated with c-myc mRNA expression (r>0.4 across all lymphoma samples) were
defined as c-myc target genes (Figure 35C). The majority of these genes have
been
previously described as c-myc target genes (Zeller 2003; Basso 2005). Stage
two
utilized the 100 genes that exhibited the largest t-statistics differentiating
expression
in BL from expression in each DLBCL subtype, excluding genes used in stage
one.
[00244] For a sample to be classified as BL, it had to be predicted to be BL
in both
stages of the predictor in each of the three pairwise comparisons between BL
and
the various DLBCL subtypes. Leave-one-out cross-validation was used to
evaluate
the predictor performance and minimize bias (Hills 1966; Ransohoff 2004;
Molinaro
2005).
[00246] Among the 25 cases identified as classic BL by pathological review,
the
gene expression-based predictor classified 100% correctly (Figure 35B).
Atypical BL
and classic BL could not be distinguished by gene expression, and therefore
the
predictor also classified 19 of the 20 atypical BL cases as BL. The cases for
which
the gene expression-based and pathology-based diagnoses were in agreement were
labeled "BL-concordant" cases. The gene expression-based predictor also
exhibited
100% accuracy in the diagnosis of ABC, GCB, and PMBL, and additionally
classified
all but one of the unclassified DLBCLs as DLBCL (Figure 35C).
[00246] In addition to leave-one-out cross-validation, the BL prediction
algorithm was
tested by dividing the cases into equally sized training and validation sets.
The
predictor was generated using data from the training set and applied to the
validation
set cases. This analysis agreed well with leave-one-out cross-validation in
99% of
the validation set cases, suggesting that the predictive algorithm is highly
effective in
distinguishing BL from DLBCL.
175
CA 2897828 2019-12-20

[00247] Having established the accuracy of the BL predictor, it was next used
to
classify those 26 cases that were originally submitted as BL or Burkitt-like
lymphoma
but were reassigned based on pathological review as either DLBCL (n=20) or
high
grade lymphoma not otherwise specified (NOS) (n=6). The expression-based
predictor disagreed with the pathological diagnosis in eight of these cases
(31%). In
addition, the expression-based predictor classified one of the nine cases
submitted
and verified as high-grade DLBCL as BL.
[00248] Altogether, nine cases that were diagnosed as either DLBCL or high
grade
lymphoma upon pathological review had gene expression profiles consistent with
BL. These cases were labeled "BL-discrepant" cases (marked by an asterisk in
Figure 35D). The BL-discrepant cases could be readily distinguished from all
subtypes of DLBCL by gene expression, and had an assigned probability of being
BL ranging from 98% to 100% (Figure 36A). The diagnosis of BL in the BL-
discrepant cases was supported by the presence of a c-myc translocation in all
cases. Four of the nine BL-discrepant cases expressed BCL2 mRNA and protein at
high levels, and three had a t(14:18) translocation involving the BCL2 gene in
addition to the 48;14) translocation. The remaining five BL-discrepant cases
were
BCL2-negative and completely indistinguishable from BL by gene expression. In
summary, although the BL-discrepant cases did not meet the WHO criteria for a
diagnosis of BL based on morphology and immunophenotype, they nonetheless
harbored a c-myc translocation and were indistinguishable from classic BL by
gene
expression.
[00249] It was next examined whether the expression-based BL predictor could
distinguish BL from DLBCL bearing a c-myc translocation. Consistent with
previous
studies, 7% of the cases submitted as DLBCL (six of the 87 cases tested) were
176
CA 2897828 2019-12-20

found to have a c-myc translocation. The gene expression profiles of these six
cases were distinct from those of BL (Figure 36B). All six cases had profiles
of
DLBCL (4 GCB, 2 ABC). Five of these cases had a BL predictor probability of
0%,
but one had a BL predictor probability of 66% and may represent a rare
biological
overlap between BL and DLBCL.
[00250] To elucidate biological mechanisms that distinguish BL from DLBCL,
hierarchical clustering (Eisen 1998) was used to organize the BL predictor
genes
according to their expression patterns across all BL and DLBCL samples. Many
of
the predictor genes segregated into four gene expression signatures reflecting
distinctive biological attributes of BL. For example, c-myc and its target
genes
constituted one gene expression signature, which was more highly expressed in
BL
than in DLBCL, reflecting the influence of the c-myc translocation in BL
(Figure 37A).
[00251] Another cluster of predictor genes that were more highly expressed in
BL
than DLBCL included genes characteristically expressed in normal GC B cells,
such
as MME (CD10) and MYBL1. This was unexpected given that GCB, like BL, is
derived from the germinal center stage of B cell differentiation (Mann 1976;
Alizadeh
2000). To define a comprehensive GC B cell gene expression signature, whole
genome microarrays were used to profile gene expression in various normal B
cell
subsets, including GC B cells, as well as resting and mitogen-stimulated blood
B
cells. GC B cell signature genes were defined as those that were overexpressed
in
GC B cells compared with the blood B cell populations, but were not associated
with
cellular proliferation. These GC B cell signature genes could be divided into
three
sets: 1) genes that were expressed more highly in BL than GCB ("BL-high"), 2)
genes that were expressed more highly expressed in GCB than BL ("BL-low"), and
177
CA 2897828 2019-12-20

3) genes that were expressed equivalently in BL and GCB (Figure 37B). Thus, BL
and GCB retain expression of different subsets of GC B cell signature genes.
[00252] A third gene expression signature included MHC class I genes that were
expressed at lower levels in BL than in DLBCL (Figure 37C). A fourth signature
included known targets of the NF-k13 pathway that were expressed at relatively
low
levels in BL. Expression of a recently defined set of NF-KB target genes (Lam
2005)
was examined, and it was found that BL expressed these genes at very low
levels
compared to each DLBCL subgroup, including GCB (Figure 37D).
[00253] Expression was quantitated for each gene expression signature within
the
various lymphoma types. For each signature, the average expression of its
component genes in a lymphoma biopsy sample was calculated to generate a gene
expression signature value for that sample. Figure 37E displays the signature
values for biopsies classified according to their molecular diagnoses. BL-
concordant
samples had signature values that were readily distinguished from those of
samples
belonging to the three DLBCL subtypes (P<1x10-7). Notably, BL-discrepant
samples
had signature values that were similar to those of BL-concordant cases, again
supporting classification as BL. The BL-discrepant cases that were BCL2-
negative
(n=5) were indistinguishable from the BL-concordant cases by gene expression.
In
contrast, the BL-discrepant cases that were BCL2-positive (n=4) had lower
expression of the BL-high GC B cell gene expression signature. The two BL-
concordant cases with a t(14;18) also had low expression of the BL-high gene
expression signature. A final important observation was that DLBCL samples
with a
c-myc translocation were easily distinguishable from BL in the expression of
all four
gene expression signatures (p<0.01).
178
CA 2897828 2019-12-20

[00254] To understand the effect of treatment on overall survival in BL,
clinical data
was analyzed from patients with a molecular diagnosis of BL for whom complete
clinical information was available. Overall survival was markedly better for
patients
who received intensive chemotherapy regimens than for those who received CHOP-
like regimens (P=0.02; Figure 38B). The inferior outcome of adult patients
treated
with CHOP-like regimens could not be accounted for by differences in age,
stage,
serum lactate dehydrogenase, performance status, sites of disease, or the
presence
of t(14;18). Among BL-discrepant cases, CHOP-like therapies were also inferior
(P=0.049), although the number of cases is low (n=7; Figure 38C). BL patients
who
received CHOP-like regiments had a lower survival compared to patients in each
of
the three DLBCL subtypes who received similar therapy (Figure 38D). Overall,
these results are consistent with previous reports (Butler 1993; Magrath 1996;
Smeland 2004) indicating that BL patients have excellent outcomes when treated
with intensive chemotherapy regimens rather than CHOP-like regimens.
Example 20: Identification and characterization of cyclin D1-neqative MCL
cases::
[00255] Lymph node biopsies from six patients with suspected cyclin Dl-
negative
MCL were examined. Two of these cases were identified in a previous study
(Rosenwald 2003a), and the other four were newly identified as cyclin D1-
negative
MCL based on immunohistochemistry and RT-PCR analysis. Each of these six
cases exhibited the characteristic morphological characteristics of MCL as
determined by a panel of expert hematopathologists.
[00256] Gene expression data was obtained using a Lymphochip microarray and
Affymetrix U133A/B microarrays, and the gene expression profile of each case
was
compared to those of ABC (78 cases), GCB (85 cases), PMBL (33 cases), FL (193
cases), MALT (14 cases), splenic marginal zone lymphoma (SMZL) (6 cases), and
179
CA 2897828 2019-12-20

SLL (14 cases) (Figure 33). The distributions of the Bayesian predictor for
each
category were used to estimate the probability that any particular sample
belonged
to that category by applying Bayes' rule (Rosenwald 2003a; Wright 2003).
Specifically, a Bayesian predictor was generated for each lymphoma category
between that category and cyclin D1-positive MCL samples, based on the 50
genes
with the largest t-statistics between them. Only those samples for which all
pair-wise
predictors agreed that there was a greater than 90% probability that the
sample was
MCL were classified as cyclin D1-negative MCL. This set of models was applied
to
the data set in a leave-one-out cross-validated fashion so that the models
tested on
a given sample were based on a data set that excluded that sample. Samples
that
exhibited less than a 90% probability of belonging to a category were deemed
"unclassified." All six suspected cyclin D1-negative MCL cases were indicated
as
MCL in all pair-wise models with at least 99.99% confidence. None of the
samples
from the other categories were indicated as MCL with greater than 20%
confidence
in their respective pair-wise comparisons. Thus, the six samples are
considered to
be bona fide cases of cyclin D1-negative MCL.
[00257] Lymph node specimens from the six cyclin D1-negative MCL cases were
fixed in 10% neutral buffered formalin and embedded in paraffin, and 4 pm
sections
were cut and stained with hematoxylin and eosin (H&E) for histologic
evaluation.
lmmunohistochemical stains for cyclin D1 protein were performed on formalin-
fixed,
paraffin-embedded tissue sections. After deparaffinization in xylene and
rehydration
in graded alcohols, endogenous peroxidase was blocked with hydrogen peroxide.
Heat-induced antigen retrieval was performed using citrate buffer, pH 6.0
(Brynes
1997). After rinsing in phosphate-buffered saline, mouse anti-cyclin D1
antibody
was applied at a dilution of 1:200 and rabbit monoclonal anti-cyclin antibody
(SP4)
180
CA 2897828 2019-12-20

(Neomarkers, Fremont, CA) was applied using the suggested procedure for
antigen
retrieval with minor modifications (Cheuk 2004) (Table 2416). Antibodies
against
CD3, CD5, CD20, CD23, CD43, cyclin 02, cyclin D3, cyclin E, retinoblastoma
protein (RB), and p27kIP1 were also employed for immunohistochemical stains
(Table
2416).
Table 2416:
Antibody Clone Source Dilution Retrieval
CD3 PS1 Ventana (Tucson, AZ) Neat A
(10 mM citrate buffer, pH
6.0, 30 min., water bath
(95 C))
CD5 4C7 Novocastra (Newcastle upon 1:20 A
Tyne, UK)
CD20 L26 DAKO (Carpinteria, CA) 1:200 A
CD23 BU38 The Binding Site (San Diego, CA) 1:5
(Protease I enzymatic
digestion, 8 min.)
CD43 L60 Ventana Neat None
Cyclin D1 DCS-6 DAKO 1:200 A
Cyclin Dl SP4 Neomarkers (Fremont, CA) 1:100
(1 mM EDTA, pH 8.0, 30
min., water bath (95 C))
Cyclin D2 Polyclonal Santa Cruz Biotech (Santa Cruz,
1:500
CA) (1 mM EDTA,
pH 8.0, 60
min., water bath (95 C))
Cyclin D3 DCS-22 Neomarkers 1:100
(10 mM citrate buffer, pH
6.0, 10 min., pressure
cooker (115 C))
Cyclin E 13A3 Novocastra 1:10
RB Rb1 DAKO 1:10 A
p27K'Pl SX53G8 DAKO 1:20 A
[00258] Stains were performed on a Ventana ES automated immunostainer
(Ventana Biotek, Tucson, AZ) with a streptavidin-biotin peroxidase detection
system.
Positivity for the cyclins, RB, and p27kiP1 was defined as a strong nuclear
staining in
more than 50% of the neoplastic cells. Results of histologic and immunologic
studies are set forth in Table 2417.
Table 2417:
Case
1 2 3 4 5 6
Growth pattern Nodular Diffuse Nodular Diffuse Nodular
Nodular
Cytology Typical Typical Typical Typical Typical Typical
CD20
181
CA 2897828 2019-12-20

CD3 - -
CD5
CD23 +(w)
Pathologic CD43 +(w)
features Cyclin D1
Cyclin D2 NA
Cyclin D3 NA
Cyclin E - NA
RB NA
p27K1P1
t(11;14)(q13;q32) - -
Genetic 11q13 (cyclin D1, Normal* Normal Normal Normal Normal
Normal
features 12p13 (cyclin D2) Normal Normal Normal Normal
Normal Normal
6p21 (cyclin D3) Normal Normal Normal Normal Normal
Normal
12p13 (p27K'P1) Normal Normal Normal Normal Normal
Normal
NA: not available; +: positive; +(w): weakly positive; -: negative; *: normal
indicates no split or
amplification
[00259] All six cases exhibited a nodular or diffuse growth pattern and
consisted of
tumor cells with typical mantle cell cytology (Figure 34A). Immunophenotypic
analysis of the tumor cells revealed a B-cell phenotype in all cases.
Expression of
CD5 antigen was noted in all six cases, while CD43 expression was observed in
five
of the six cases. The tumor cells in one of the six cases were weakly positive
for
CD23 antigen.
[00260] All six cases were negative for cyclin D1 using the mouse monoclonal
antibody DCS-6 (Figure 34B). All four of the cases that were tested using the
rabbit
monoclonal antibody SP4 were negative for cyclin D1. Two cases (1 and 2)
demonstrated overexpression of cyclin D2 by immunostaining (Figure 34C), which
correlated well with the increased cyclin D2 mRNA levels detected by
microarray
analysis (Figure 33, lower panel). Both of these cases were negative for
cyclin D3.
Three cases (3-5) exhibited overexpression of cyclin D3 by immunostaining
(Figure
34D), which correlated well with the increased cyclin D3 mRNA levels detected
by
microarray analysis (Figure 33, lower panel). Case 6 also showed upregulation
of
cyclin D3 mRNA by microarray analysis (Figure 33, lower panel), but the tissue
block
for this case was not available for immunostaining.
182
CA 2897828 2019-12-20

[002611A11 six cases were negative for cyclin E, but showed positive
immunostaining
for RB. RB expression levels were similar in all of the cases, and were
comparable
to those seen in cyclin D1-positive MCL. Downregulation of p271(1Pl expression
was
observed in all six cases, with the intensity of nuclear staining much weaker
than
that seen in reactive T-lymphocytes.
[00262] lnterphase FISH analysis was performed on cells left over from prior
cytogenetic analyses or on formalin-fixed, paraffin-embedded tissue sections.
For
detection of the t(11;14)(q13;q32), a commercially-available LSI IGH/CCND1
double-color, double-fusion probe was used (Vysis Inc., Downers Grove, IL).
[00263] For break-apart FISH assays for the CCND1 (11q13), CCND2 (12p13), and
CDKN1B/p27KIP1 (12p13) loci, appropriate BAC clones flanking the respective
genes were selected using bioinformatic resources available from the
University of
California at Santa Cruz. All BAC clones were derived from the RPCI11 library
and
were obtained from lnvitrogen/Research Genetics or the Sanger Center. The
following clones were used: CCND1 (pooled RP11-211G23/RP11-378E8 and pooled
RP11-30016/RP11-626H12), CCND2 (RP11-578L13 and RP11-388F6), and
CDKN1B/p27KIP1 (RP11-180M15 and RP11-59H1). For each locus, centromeric
and telomeric BAC clones were differentially labeled with Spectrum Orange or
Spectrum Green (Vysis Inc.) and pooled for break-apart assays. Bacterial
culture,
BAC DNA isolation and labeling, probe preparation, and FISH on cytogenetic
suspensions were performed as previously described (Schlegelberger 1999;
Martin-
Subero 2002). The CCND3 locus was investigated using a recently-described
break-apart assay (Sonoki 2001). Locus-specific interphase FISH was performed
on
paraffin-embedded tissue sections according to the manufacturer's instructions
183
CA 2897828 2019-12-20

(Vysis Inc.), or recently-described protocols (Ye 2003) with minor
modifications.
Whenever possible, at least 100 cells were analyzed.
[00264] None of the six cases displayed the IGH/CCND1 fusion. FISH studies
with
the locus-specific probe were also negative for variant translocations or
amplifications involving the CCND1 locus at band 11q13 in all six cases (Table
2417). Conventional cytogenetic analysis was also performed on case 6, and did
not reveal a chromosomal alteration affecting band 11q13. FISH analysis using
break-apart probes for the CCND1 (12p13), CCND2 (6p21), and CDKN1B/p27KIP1
(12p13) loci did not reveal any evidence of chromosomal translocation or
amplification in these cases (Table 2417).
[00265] The INK4a/ARF locus encodes the tumor suppressor proteins p16INK4a and
p 1 4AR F. To detect genomic loss of the INK4a/ARF tumor suppressor locus in
the
specimens, quantitative real-time PCR assays were performed using genomic DNA
as previously described (Rosenwald 2003a). The REL locus on chromosome 2p
was chosen as the reference gene, and a cutoff ratio of INK4a/ARF locus copy
number relative to REL locus copy number was used to assess tumor DNA for
genomic deletions. A tumor DNA sample that yielded an INK4a/ARF to REL ratio
below the cutoff ratio was considered to have a genomic deletion of the
INK4a/ARF
locus. The primers and probe sets for the INK4a/ARF and the REL loci have been
described previously (Goff 2000; Labuhn 2001). No INK4A/ARF locus deletions
were detected in any of the six cases.
[00266] Clinical features of the six patients are summarized in Table 2418.
The
patients consisted of five males and one female with a median age of 61 years
(range, 54-77 years). All patients presented with stage IV disease.
Lymphadenopathy was the most common presentation and extranodal sites were
184
CA 2897828 2019-12-20

involved by lymphoma in all six patients. Five patients received combination
chemotherapy initially, but none of these patients achieved a complete
clinical
response. One patient (case 1) was not treated initially and developed
gastrointestinal involvement 26 months after the initial diagnosis. At the
time of last
follow-up, one of the patients had died and the other five were alive with
disease.
[00267] Ninety-three cases (92 cases in the original study plus the false
negative
case) of cyclin Dl-positive MCL (Rosenwald 2003a) were used for comparison.
The
median follow-up for these patients was 26 months (range, 7-166 months). The
clinical features including age and sex distribution, stage, presence of B
symptoms,
serum lactate dehydrogenase (LDH) levels, extranodal sites, IPI scores, types
of
treatment, and clinical responses were similar between the cyclin D1-positive
and
cyclin D1-negative groups. At the time of last follow-up, 65 of 93 patients
with cyclin
D1-positive MCL had died, with a median overall survival of 31 months. No
significant difference in overall survival was identified between the cyclin
D1-positive
and cyclin D1-negative groups.
Table 2418:
Case
1 2 3 4 5 6
Age (years)/sex 54/F 611M 61/M 60/M 54/M 77/M
Ann Arbor stage IV IV IV IV IV IV
B symptoms + -
Serum LDH levels Normal High Normal High Normal Normal
Extranodal sites BM, PB BM BM BM, spleen BM BM, lung,
GI
IPI score 2 3 2 3 2 3
Initial therapy None R-CHOP CHOP COP CHOP COP
Response NA PR PR PR PR PR
Progression
Follow-up 38 5 88 19 70 30
(months)
Status AWD AWD DOD AWD AWD AWD
LDH, lactate dehydrogenase; BM, bone marrow; PB, peripheral blood; GI,
gastrointestinal tract; R,
Rituxan; PR, partial response; AWD, alive with disease; DOD, dead of disease
[00268]As stated above, the foregoing is merely intended to illustrate various
embodiments of the present invention. The specific modifications discussed
above
185
CA 2897828 2019-12-20

are not to be construed as limitations on the scope of the invention. It will
be
apparent to one skilled in the art that various equivalents, changes, and
modifications may be made without departing from the scope of the invention,
and it
is understood that such equivalent embodiments are to be included herein
REFERENCES
1. Alizadeh, A.A., et al. 1998. Probing lymphocyte biology by genomic-scale
gene expression analysis. J Clin Immunol 18:373-79.
2. Alizadeh, A.A., et al. 1999. The Lymphochip: a specialized cDNA
microarray
for the genomic-scale analysis of gene expression in normal and malignant
lymphocytes. Cold Spring Harbor Symp Quant Biol 64:71-78.
3. Alizadeh, A.A., et al. 2000. Distinct types of diffuse large B-cell
lymphoma
identified by gene expression profiling. Nature 403:503-511.
4. Alon, U., et al. 1999. Broad patterns of gene expression revealed by
clustering analysis of tumor and normal colon tissues probed by
oligonucleotide
arrays. Proc Natl Acad Sci USA 96:6745-6750.
5. Andreasson, P., et al. 1998. Genomic amplifications of CCDN2 is rare in
non-
Hodgkin lymphomas. Cancer Genet Cytogenet 102:81-82.
6. Basso, K., et al. 2004. Tracking CD40 signaling during germinal center
development. Blood 104:4088-4096.
7. Basso, K., et al. 2005. Reverse engineering of regulatory networks in
human
B cells. Nat Genet 37:382-390.
8. Bayes, T. 1763. An essay towards solving a problem in the doctrine of
chances. Phil Trans Roy Soc London 53:370.
186
CA 2897828 2019-12-20

9. Bea, S., et al. 1999. Increased number of chromosomal imbalances and
high-
level DNA amplifications in mantle cell lymphoma are associated with blastoid
variants. Blood 93:4365-4374.
10. Bea, S., et al. 2004. Clinicopathologic significance and prognostic
value of
chromosomal imbalances in diffuse large B-cell lymphomas. J Clin Oncol 22:3498-
3506.
11. Bea, S., et al. 2005. Diffuse large B-cell lymphoma subgroups have
distinct
genetic profiles that influence tumor biology and improve gene expression-
based
survival prediction. Blood 106:3183-3190.
12. Berglund, M., et al. 2002. Chromosomal imbalances in diffuse large B-
cell
lymphoma detected by comparative genomic hybridization. Mod Pathol 15:807-816.
13. Bergsagel, P.L., et al. 2003. Critical roles for immunoglobulin
translocations
and cyclin D dysregulation in multiple myeloma. Immunol Rev 194:96-104.
14. Bishop, P.C., Rao, V.K., Wilson, W.H. 2000. Burkitt's lymphoma:
molecular
pathogenesis and treatment. Cancer Invest 18:574-583.
15. Boxer, L.M., Lozanski, G., Byrd, J.C. 2001. Translocations involving c-
myc
and c-myc function. Oncogene 20:5595-5610.
16. Brynes, R.K., et al. 1997. Demonstration of cyclin D1 (bcl-1) in mantle
cell
lymphoma. Enhanced staining using heat and ultrasound epitope retrieval. App!
lmmunohistochem 5:45-48.
17. Butler, R.D., Hainsworth, J.D. 1993. Optimal therapy for small
noncleaved cell
lymphoma. Cancer Treat Res 66:65-79.
18. Chee, M., et al. 1996. Accessing genetic information with high density
DNA
arrays. Science 274:610-14.
187
CA 2897828 2019-12-20

19. Cheuk, W., et at. 2004. Consistent immunostaining for cyclin D1 can be
achieved on a routine basis using a newly available rabbit monoclonal
antibody. Am
J Surg Pathol 28:801-807.
20. Chiarle, R., et at. 2000. Increased proteasome degradation of cyclin-
dependent kinase inhibitor p27 is associated with a decreased overall survival
in
mantle cell lymphoma. Blood 95:619-626.
21. Cho, R.J., et at. 1998. A genome-wide transcriptional analysis of the
mitotic
cell cycle. Mol Cell 2:65-73.
22. Chu, S., et at. 1998. The transcriptional program of sporulation in
budding
yeast. Science 282:699-705.
23. Ciemerych, M.A., et al. 2002. Development of mice expressing a single D-
type cyclin. Genes Dev 16:3277-3289.
24. Cigudosa, J.C., et at. 1999. Cytogenetic analysis of 363 consecutively
ascertained diffuse large B-cell lymphomas. Genes Chromosomes Cancer 25:123-
133.
25. Copie-Bergman, C., et at. 2002. MAL expression in lymphoid cells:
further
evidence for MAL as a distinct molecular marker of primary mediastinal large 6-
cell
lymphomas. Mod Pathol 15:1172-1180.
26. Copie-Bergman, C., et al. 2003. Interleukin 4-induced gene us activated
in
primary mediastinal large B-cell lymphoma. Blood 101:2756-2761.
27. Dave, B.J., et at. 2002. Cytogenetic characterization of diffuse large
cell
lymphoma using multi-color fluorescence in situ hybridization. Cancer Genet
Cytogenet 132:125-132.
28. Delmer, A., et at. 1995. Overexpression of cyclin D2 in chronic B-cell
malignancies. Blood 85:2870-2876.
188
CA 2897828 2019-12-20

29. DeRisi, J., et al. 1996. Use of a cDNA microarray to analyze gene
expression
patterns in human cancer. Nat Genet 14:457-60.
30. DeRisi, J.L., lyer, V.R., Brown, P.O. 1997. Exploring the metabolic and
genetic control of gene expression on a genomic scale. Science 278:680-86.
31. Divine, M., et al. 2005. Burkitt lymphoma in adults: a prospective
study of 72
patients treated with an adapted pediatric LMB protocol. Ann Oncol 16:1928-
1935.
32. Doglioni, C., et al. 1998. Cyclin D3 expression in normal, reactive and
neoplastic tissues. J Pathol 185:159-166.
33. Drapner, H. 1966. Applied regression. Wiley, New York.
34. Dudoit, S., Fridlyand, J., Speed, T.P. 2002. Comparison of
discrimination
methods for the classification of tumors using gene expression data. J Am Stat
Assoc 97:77-87.
35. Eisen, M.B., Spellman, P.T., Brown, P.O., Botstein, D. 1998. Cluster
analysis
and display of genome-wide expression patterns. Proc Natl Acad Sci USA
95:14863-
14868.
36. Feuerhake, F., et al. 2005. NFkappaB activity, function, and target-
gene
signatures in primary mediastinal large B-cell lymphoma and diffuse large B-
cell
lymphoma subtypes. 106:1392-1399.
37. Fisher, R.I., et al. 1993. Comparison of a standard regimen (CHOP) with
three intensive chemotherapy regimens for advanced non-Hodgkin's lymphoma. N
Engl J Med 328:1002-1006.
38. Furey, T.S., et al. 2000. Support vector machine classification and
validation
of cancer tissue samples using microarray expression data. Bioinformatics
16:906-
914.
189
CA 2897828 2019-12-20

39. Gerbitz, A., et at. 1999. Deregulation of the proto-oncogene c-myc
through
t(8;22) translocation in Burkitt's lymphoma. Oncogene 18:1745-1753.
40. Goff, L.K., et at. 2000. The use of real-time quantitative polymerase
chain
reaction and comparative genomic hybridizations to identify amplification of
the REL
gene in follicular lymphoma. Br J Haematol 111:618-625.
41. Golub, T.R., et at. 1999. Molecular classification of cancer: class
discovery
and class prediction by gene expression monitoring, Science 286:531-537.
42. Gress, T.M., et at. 1996. A pancreatic cancer-specific expression
profile.
Oncogene 13:1819-30.
43. Haralambieva, E., et at. 2005. Clinical, immunophenotypic, and genetic
analysis of adult lymphomas with morphologic features of Burkitt lymphoma. Am
J
Surg Pathol 29:1086-1094.
44. Harris, N.L., et at. 1994. A revised European-American classification
of
lymphoid neoplasms: a proposal from the International Lymphoma Study Group.
Blood 84:1361-1392.
45. Hashimoto, Y., et at. 2002. The evaluation of the biological behavior
and
grade among cases with mantle cell lymphoma. Leuk Lymphoma 43:523-530.
46. Hecht, J.L., et at. 2000. Molecular biology of Burkitt's lymphoma. J
Clin Oncol
18:3707-3721.
47. Heller, R.A., et at. 1997. Discovery and analysis of inflammatory
disease-
related genes using cDNA microarrays. Proc Natl Acad Sci USA 94:2150-55.
48. Hills, M. 1966. Allocation rules and error rates. J Royal Statis Soc
[B] 28:1-31.
49. Holstege, F.C., et al. 1998. Dissecting the regulatory circuitry of a
eukaryotic
genome. Cell 95:717-728.
190
CA 2897828 2019-12-20

50. Huang, J.Z., et al. 2002. The t(14;18) defines a unique subset of
diffuse large
B-cell lymphoma with a germinal center B-cell gene expression profile. Blood
99:2285-2290.
51. Hyman, E., et al. 2002. Impact of DNA amplification on gene expression
patterns in breast cancer. Cancer Res 62:6240-6245.
52. lqbal, J., et al. 2004. BCL2 translocation defines a unique tumor
subset within
the germinal center B-cell-like diffuse large B-cell lymphoma. Am J Pathol
165:159-
166.
53. Irizarry, R.A., et al. 2003. Exploration, normalization, and summaries
of high
density oligonucleotide array probe level data. Biostatistics 4:249-264.
54. Hills, M. 1966. Allocation rules and error rates. J Royal Statis Soc
Series B
28:1-31.
55. Inaba, T., et al. 1992. Genomic organization, chromosomal localization,
and
independent expression of human cyclin D genes. Genomics 13:565-574.
56. Jaffe, E.S., Harris, N.L., Stein, H., Vardiman, J.W. 2001. Tumors of
hematopoietic and lymphoid tissues. IARC Press, Lyon.
57. Jares, P, et al. 1996. Expression of retinoblastoma gene product (pRb)
in
mantle cell lymphomas. Correlation with cyclin D1 (PRAD1/CCND1) mRNA levels
and proliferative activity. Am J Pathol 148:1591-1600.
58. Khouri, I.F., et al. 1998. Hyper-CVAD and high-dose
methotrexate/cytarabine
followed by stem-cell transplantation: an active regimen for aggressive mantle-
cell
lymphoma. J Clin Oncol 12:3803-3809.
59. Kohonen, T. 1997. Self-organizing maps. Springer Press, Berlin.
60. Kramer, M.H., et al. 1998. Clinical relevance of BCL2, BCL6, and MYC
rearrangements in diffuse large B-cell lymphoma. Blood 92:3152-3162.
191
CA 2897828 2019-12-20

61. Labuhn, M., et al. 2001. Quantitative real-time PCR does not show
selective
targeting of p14(ARF) but concomitant inactivation of both p16(INK4A) and
p14(ARF) in 105 human primary gliomas. Oncogene 20:1103-1109.
62. Lam, L.T., et al. 2005. Small molecular inhibitors of IkB-kinase are
selectively
toxic for subgroups of diffuse large B cell lymphoma defined by gene
expression
profiling. Clin Cancer Res 11:28-40.
63. Lashkari, D.A., et al. 1997. Yeast microarrays for genome wide parallel
genetic and gene expression analysis. Proc Natl Acad Sci USA 94:13057-62.
64. Li, C., Wong, W.H. 2001. Model-based analysis of oligonucleotide
arrays:
expression index computation and outlier detection. Proc Natl Acad Sci USA
98:31-
36.
65. Lin, Z., et al. 2003. Growth regulation by p27K1p1 is abrogated by
multiple
mechanisms in aggressive malignant lymphomas. Br J Haematol 121:739-748.
66. Lipshutz, R.J., et al. 1995. Using oligonucleotide probe arrays to
access
genetic diversity. Biotechniques 19:442-47.
67. Lockhart, D.J., et al. 1996. Expression monitoring by hybridization to
high-
density oligonucleotide arrays. Nat Biotechnol 14:1675-80.
68. Macpherson, N., et al. 1999. Small noncleaved, non-Burkitt's (Burkitt-
Like)
lymphoma: cytogenetics predict outcome and reflect clinical presentation. J
Clin
Oncol 17:1558-1567.
69. Magrath, E.S., et al. 2001. Tumours of Haematopoietic and Lymphoid
Tissues. Lyon: IARC Press.
70. Mann, R.B., et al. 1976. Non-endemic Burkitt's lymphoma. A B-cell tumor
related to germinal centers. N Engl J Med 295:685-691.
192
CA 2897828 2019-12-20

71. Martin-Subero, J.I., et al. 2002. Multicolor-FICTION: expanding the
possibilities of combined morphologic, immunophenotypic, and genetic single
cell
analyses. Am J Pathol 161:413-420.
72. Mead, G.M., et al. 2002. An international evaluation of CODOX-M and
CODOX-M alternating with IVAC in adult Burkitt's lymphoma: results of United
Kingdom Lymphoma Group LY06 study. Ann Oncol 13:1264-1274.
73. Molinaro, A.M., Simon, R., Pfeiffer, R.M. 2005. Predictor error
estimation: a
comparison of resampling methods. Bioinformatics 21:3301-3307.
74. Monni, 0., et al. 1996. DNA copy number changes in diffuse large B-cell
lymphoma-comparative genomic hybridization study. Blood 87:5269-5278.
75. Morton, L.M., et al. 2005. Lymphoma incidence patterns by WHO subtype
in
the United States, 1992-2001. Blood 107:265-276.
76. Nanjangud, G., et al. 2002. Spectral karyotyping identifies new
rearrangements, translocations, and clinical associations in diffuse large B-
cell
lymphoma. Blood 99:2554-2561.
77. Neri, A., et al. 1988. Different regions of the immunoglobulin heavy-
chain
locus are involved in chromosomal translocations in distinct pathogenetic
forms of
Burkitt lymphoma. Proc Natl Acad Sci USA 85:2748-2752.
78. Orsetti, B., et al. 2004. Genomic and expression profiling of
chromosome 17
in breast cancer reveals complex patterns of alterations and novel candidate
genes.
Cancer Res 64:6453-6460.
79. Ott, MM., et al. 1997. Cyclin D1 expression in mantle cell lymphoma is
accompanied by downregulation of cyclin D3 and is not related to the
proliferative
activity. Blood 90:3154-3159.
193
CA 2897828 2019-12-20

80. Pease, A.G., et al. 1994. Light generated oligonucleotide arrays for
rapid DNA
sequence analysis. Proc Natl Acad Sci USA 91:5022-26.
81. Pees, H.W., et al. 1992. The BFM-protocol for HIV-negative Burkitt's
lymphomas and L3 ALL in adult patients: a high chance for cure. Ann Hematol
65:201-205.
82. Phillips, J.L., et al. 2001. The consequences of chromosomal aneuploidy
on
gene expression profiles in a cell line model for prostate carcinogenesis.
Cancer Res
61:8143-8149.
83. Pietu, G., et al. 1996. Novel gene transcripts preferentially expressed
in
human muscles revealed by quantitative hybridization of a high density cDNA
array.
Genome Res 6:492-503.
84. Pollack, J.R., et al. 2002. Microarray analysis reveals a major direct
role of
DNA copy number alteration in the transcriptional program of human breast
tumors.
Proc Natl Acad Sci USA 99:12963-12968.
85. Polyak, K., et al. 1994. p27Kip1, a cyclin-Cdk inhibitor, links
transforming
growth factor-beta and contact inhibition to cell cycle arrest. Genes Dev 8:9-
22.
86. Pruneri, G., et al. 2003. lmmunoreactivity for cyclin D3 is frequently
detectable in high-grade primary gastric lymphomas in the absence of the
46;14)(p21.1;q32.3) chromosomal translocation. J Pathol 200:596-601.
87. Quintanilla-Martinez, L., et al. 1998. Mantle cell lymphomas lack
expression
of p27Kip1, a cyclin-dependent kinase inhibitor. Am J Pathol 153:175-182.
88. Quintanilla-Martinez, L., et al. 2003. Sequestration of p27Kip1 protein
by
cyclin D1 in typical and blastic variants of mantle cell lymphoma (MCL):
implications
for pathogenesis. Blood 101:3181-3187.
194
CA 2897828 2019-12-20

89. Radmacher, M.D., McShane, L.M., Simon, R. 2002. A paradigm for class
prediction using gene expression profiles. J Comput Biol 9:505-511.
90. Ramaswamy, S., et al. 2001. Multidass cancer diagnosis using tumor gene
expression signatures. Proc Nat! Acad Sci USA 98:15149-15154.
91. Ransohoff, D.F. 2004. Rules of evidence for cancer molecular-marker
discovery and validation. Nat Rev Cancer 4:309-314.
92. Rao, P.H., et al. 1998. Chromosomal and gene amplification in diffuse
large
B-cell lymphoma. Blood 92:234-240.
93. Rosenberg, C.L., et al. 1991. PRAD1, a candidate BCL1 oncogene: mapping
and expression in centrocytic lymphoma. Proc Nat! Acad Sci USA 88:9638-9642.
94. Rosenwald, A., et al. 2002. The use of molecular profiling to predict
survival
after chemotherapy for diffuse large-B-cell lymphoma. New Engl J Med 346:1937-
1947.
95. Rosenwald, A., et al. 2003a. The proliferation gene expression
signature is a
quantitative integrator of oncogenic events that predicts survival in mantle
cell
lymphoma. Cancer Cell 3:185-197.
96. Rosenwald, A., et al. 2003b. Molecular diagnosis of primary mediastinal
B
cell lymphoma identifies a clinically favorable subgroup of diffuse large B
cell
lymphoma related to Hodgkin lymphoma. J Exp Med 198:851-862.
97. Savage, K.J., et al. 2003. The molecular signature of mediastinal large
B-cell
lymphoma differs from that of other diffuse large B-cell lymphomas and shares
features with classical Hodgkin lymphoma. Blood 102:3871-3879.
98. Schena, M., Shalon, D., Davis, R.W., Brown, P.O. 1995. Quantitative
monitoring of gene expression patterns with a complementary DNA microarray.
Science 270:467-70.
195
CA 2897828 2019-12-20

99. Schena, M.,
et al. 1996. Parallel human genome analysis: rnicroarray based
expression monitoring of 1000 genes. Proc Natl Acad Sci USA 93:10614-19.
100. Schlegelberger, B., et al. 1999. Classical and molecular cytogenetics of
tumor
cells. In: Diagnostic Cytogenetics. Springer-Verlag, Berlin, Heidelberg; pp.
151-185.
101. Shaffer, A.L., et al. 2001. Signatures of the immune response. Immunity
15:375-385.
102. Shalon, D., Smith, S.J., Brown, P.O. 1996. A DNA microarray system for
analyzing complex DNA samples using two-color fluorescent probe hybridization.
Genome Res 6:639-45.
103. Sherr, C.J., et at. 1994. D-type cyclins and their cyclin-dependent
kinases: G1
phase integrators of the mitogenic response. Cold Spring Harb Symp Quant Blot
59:11-19.
104. Sherr, C.J. 1996. Cancer cell cycles. Science 274:1672-1677.
105. Sherr, C.J., McCormick, F. 2002. The RB and p53 pathways in cancer.
Cancer Cell 2:103-112.
106. Shipp, M.A., et al. 2002. Diffuse large B-cell lymphoma outcome
prediction by
gene-expression profiling and supervised machine learning. Nat Med 8: 68-74.
107. Sicinski, P., et at. 1995. Cyclin D1 provides a link between development
and
oncogenesis in the retina and breast. Cell 82:621-630.
108. Sicinska, E., et al. 2003. Requirement for cyclin D3 in lymphocyte
development and T cell leukemias. Cancer Cell 4:451-461.
109. Sicinski, P., et at. 1996. Cyclin D2 is an FSH-responsive gene involved
in
gonadal cell proliferation and oncogenesis. Nature 384:470-474.
110. Simon, R.M., et at. 2003. Design and Analysis of DNA Microarray
Investigations. Springer-Verlag, New York.
196
CA 2897828 2019-12-20

111. Smeland, S., et at. 2004. Treatment of Burkitt's/Burkitt-like lymphoma in
adolescents and adults: a 20-year experience from the Norwegian Radium
Hospital
with the use of three successive regimens. Ann Oncol 15:1072-1078.
112. Sonoki, T, et at. 2001. Cyclin D3 is a target gene of
t(6;14)(p21.1;q32.3) of
mature B-cell malignancies. Blood 98:2837-2844.
113. Soussain, C., et al. 1995. Small noncleaved cell lymphoma and leukemia in
adults. A retrospective study of 65 adults treated with the LMB pediatric
protocols.
Blood 85:664-674.
114. Southern, E.M., Maskos, U., Elder, J.K. 1992. Analyzing and comparing
nucleic acid sequences by hybridization to arrays of oligonucleotides:
evaluation
using experimental models. Genomics 13:1008-17.
115. Southern, E.M., et al. 1994. Arrays of complementary oligonucleotides for
analysing the hybridisation behaviour of nucleic acids. Nucl Acids Res 22:1368-
73.
116. Spellman, PT., et at. 1998. Comprehensive identification of cell cycle
regulated genes of the yeast Saccharomyces cerevisiae by microarray
hybridization.
Mol Biol Cell 9:3273-3297.
117. Suzuki, R., et al. 1999. Selective usage of D-type cyclins in lymphoid
malignancies. Leukemia 13:1335-1342.
118. Tamayo, P., et at. 1999. Interpreting patterns of gene expression with
self-
organizing maps: methods and application to hematopoietic differentiation.
Proc Natl
Acad Sci USA 96:2907-2912.
119. Tavazoie, S., etal. 1999. Systematic determination of genetic network
architecture. Nat Genet 22:281-285.
120. Teramoto, N., et at. 1999. Expression of cyclin D2 and D3 in lymphoid
lesions. Int J Cancer 81:543-550.
197
CA 2897828 2019-12-20

121. Thomas, D.A., et al. 1999. Hyper-CV AD program in Burkitt's-type adult
acute
lymphoblastic leukemia. J Clin Oncol 17:2461-2470.
122. Tibshirani, R., Hastie, T., Narasimhan, B., Chu, G. 2002. Diagnosis of
multiple
cancer types by shrunken centroids of gene expression. Proc Natl Acad Sci USA
99:6567-6572.
123. Velculescu, V.E., Zhang, L., Vogelstein, B., Kinzler, K.W. 1995. Serial
analysis of gene expression. Science 270:484-87.
124. Virtaneva, K., et al. 2001. Expression profiling reveals fundamental
biological
differences in acute myeloid leukemia with isolated trisomy 8 and normal
cytogenetics. Proc Natl Acad Sci USA 98:1124-1129.
125. Voltz, R., Jilg, W., Wolf, H. 1989. Modification of HLA expression as a
possible factor in the pathogenesis of Burkitt's lymphoma. Haematol Blood
Transfus
32:289-292.
126. Vose, J.M., et al. 2002. CNOP for diffuse aggressive non-Hodgkin's
lymphoma: the Nebraska lymphoma study group experience. Leuk Lymphoma
43:799-804.
127. Westfall, P.H., Young, S.S. 1993. Resampling-based Multiple Testing.
Wiley,
New York.
128. Wodicka, L., et al. 1997. Genome-wide expression monitoring in
Saccharomyces cerevisiae. Nat Biotechnol 15:1359-6714.
129. Wright, G., et al. 2003. A gene expression-based method to diagnose
clinically distinct subgroups of diffuse large B cell lymphoma. Proc Natl Acad
Sci
USA 100:9991-9996.
130. Yang, W.I., et al. 1994. Cyclin D1 (BcI-1, PRAD1) protein expression in
low-
grade B-cell lymphomas and reactive hyperplasia. Am J Pathol 145:86-96.
198
CA 2897828 2019-12-20

131. Yatabe, Y., et al. 2000. Significance of cyclin D1 overexpression for the
diagnosis of mantle cell lymphoma: a clinicopathologic comparison of cyclin D1-
positive MCL and cyclin D1-negative MCL-like B-cell lymphoma. Blood 95:2253-
2261.
132. Ye, H, et al. 2003. Variable frequencies of t(11;18)(q21;q21) in MALT
lymphomas of different sites: significant association with CagA strains of H
pylori in
gastric MALT lymphoma. Blood 102:1012-1018.
133. Yunis, J.J., et al. 1989. bc1-2 and other genomic alterations in the
prognosis of
large-cell lymphoma. N Engl J Med 320:1047-1054.
134. Zeller, K.I., et al. 2003. An integrated database of genes responsive to
the
Myc oncogenic transcription factor: identification of direct genomic targets.
Genome
Biol 4:R69.
199
CA 2897828 2019-12-20

Representative Drawing
A single figure which represents the drawing illustrating the invention.
Administrative Status

2024-08-01:As part of the Next Generation Patents (NGP) transition, the Canadian Patents Database (CPD) now contains a more detailed Event History, which replicates the Event Log of our new back-office solution.

Please note that "Inactive:" events refers to events no longer in use in our new back-office solution.

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Event History , Maintenance Fee  and Payment History  should be consulted.

Event History

Description Date
Maintenance Request Received 2024-07-24
Maintenance Fee Payment Determined Compliant 2024-07-24
Letter Sent 2022-11-21
Offer of Remission 2022-11-21
Remission Not Refused 2022-11-21
Letter Sent 2022-10-21
Offer of Remission 2022-10-21
Inactive: Grant downloaded 2022-06-23
Letter Sent 2022-06-21
Grant by Issuance 2022-06-21
Inactive: Cover page published 2022-06-20
Pre-grant 2022-02-16
Inactive: Final fee received 2022-02-16
Notice of Allowance is Issued 2021-10-22
Notice of Allowance is Issued 2021-10-22
Letter Sent 2021-10-22
Inactive: IPC deactivated 2021-10-09
Inactive: IPC deactivated 2021-10-09
Inactive: Approved for allowance (AFA) 2021-08-04
Inactive: QS passed 2021-08-04
Amendment Received - Voluntary Amendment 2021-07-15
Amendment Received - Voluntary Amendment 2021-07-15
Examiner's Interview 2021-06-23
Inactive: Adhoc Request Documented 2021-06-18
Withdraw from Allowance 2021-06-18
Inactive: Approved for allowance (AFA) 2021-05-27
Inactive: Q2 passed 2021-05-27
Amendment Received - Voluntary Amendment 2020-11-12
Common Representative Appointed 2020-11-08
Inactive: COVID 19 - Deadline extended 2020-07-16
Examiner's Report 2020-07-13
Inactive: Report - QC passed 2020-07-08
Inactive: IPC deactivated 2020-02-15
Amendment Received - Voluntary Amendment 2019-12-20
Common Representative Appointed 2019-10-30
Common Representative Appointed 2019-10-30
Inactive: S.30(2) Rules - Examiner requisition 2019-06-26
Inactive: Report - QC passed 2019-06-25
Inactive: IPC assigned 2019-06-07
Inactive: IPC assigned 2019-06-07
Inactive: IPC assigned 2019-06-07
Inactive: IPC assigned 2019-06-07
Inactive: IPC assigned 2019-06-07
Inactive: First IPC assigned 2019-06-07
Inactive: IPC expired 2019-01-01
Inactive: IPC expired 2019-01-01
Letter Sent 2018-12-20
Change of Address or Method of Correspondence Request Received 2018-01-12
Inactive: IPC expired 2018-01-01
Letter sent 2017-06-23
Correct Applicant Request Received 2017-05-17
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Correct Applicant Requirements Determined Compliant 2016-08-10
Inactive: Applicant deleted 2016-08-10
Inactive: Applicant deleted 2016-08-10
Inactive: Applicant deleted 2016-08-10
Inactive: Applicant deleted 2016-08-10
Inactive: Applicant deleted 2016-08-10
Inactive: Applicant deleted 2016-08-10
Inactive: Applicant deleted 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Letter Sent 2016-08-10
Inactive: Office letter 2016-08-10
Inactive: Applicant deleted 2016-08-08
Inactive: Multiple transfers 2016-07-19
Inactive: Office letter 2015-12-31
Inactive: <RFE date> RFE removed 2015-12-31
Inactive: Cover page published 2015-09-21
Letter Sent 2015-09-16
Letter Sent 2015-09-16
Letter Sent 2015-09-16
Letter Sent 2015-09-16
Letter Sent 2015-09-16
Letter Sent 2015-09-16
Letter Sent 2015-09-16
Letter Sent 2015-09-16
Letter Sent 2015-09-16
Letter Sent 2015-09-16
Divisional Requirements Determined Compliant 2015-09-11
Inactive: Divisional - Presentation date updated 2015-09-10
Letter Sent 2015-09-10
Correct Applicant Requirements Determined Compliant 2015-09-10
Application Received - Divisional 2015-09-10
Inactive: Single transfer 2015-08-14
Inactive: Reply to s.37 Rules - Non-PCT 2015-08-14
Inactive: Correspondence - Formalities 2015-08-14
Inactive: Office letter 2015-07-27
Inactive: IPC assigned 2015-07-24
Inactive: First IPC assigned 2015-07-24
Inactive: IPC assigned 2015-07-24
Inactive: IPC removed 2015-07-24
Inactive: IPC assigned 2015-07-24
Inactive: IPC assigned 2015-07-24
Application Received - Regular National 2015-07-23
Inactive: QC images - Scanning 2015-07-21
Request for Examination Requirements Determined Compliant 2015-07-21
All Requirements for Examination Determined Compliant 2015-07-21
Inactive: Pre-classification 2015-07-21
Application Published (Open to Public Inspection) 2008-01-31

Abandonment History

There is no abandonment history.

Maintenance Fee

The last payment was received on 2021-07-16

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
THE UNITED STATES OF AMERICA, REPRESENTED BY THE SECRETARY, DEPARTMENT O
THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMEN
BOARD OF REGENTS OF THE UNIVERSITY OF NEBRASKA
UNIVERSITY OF ROCHESTER
QUEEN MARY AND WESTFIELD COLLEGE, UNIVERSITY OF LONDON
BRITISH COLUMBIA CANCER AGENCY BRANCH
OSLO UNIVERSITY HOSPITAL HF
ARIZONA BOARD OF REGENTS ON BEHALF OF THE UNIVERSITY OF ARIZONA
INSTITUT CATALA D'ONCOLOGIA
UNIVERSITAT DE BARCELONA
FUNDACIO CLINIC
HOSPITAL CLINIC
JULIUS-MAXIMILIANS-UNIVERSITY OF WURZBURG
Past Owners on Record
ANDREAS ROSENWALD
ANDREAS ZETTL
ANDREW LISTER
ARMANDO LOPEZ-GUILLERMO
BRUCE TAN
DENNIS WEISENBURGER
ELAINE JAFFE
ELIAS CAMPO GUERRI
EMILIO MONTSERRAT COSTA
ERLEND SMELAND
GEORGE WRIGHT
GERMAN OTT
HANS-KONRAD MULLER-HERMELINK
HARALD HOLTE
ITZIAR SALAVERRIA
JAMES ARMITAGE
JAN DELABIE
JOHN I. POWELL
JOSEPH CONNORS
JULIE VOSE
KAI FU
LISA RIMSZA
LOUIS M. STAUDT
RANDY GASCOYNE
RICHARD I. FISHER
SANDEEP DAVE
SILVIA M. BEA
STEIN KVALOY
THOMAS GROGAN
THOMAS MILLER
TIMOTHY C. GREINER
VICTOR MORENO
WING C. CHAN
WYNDHAM WILSON
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2015-07-20 200 9,769
Abstract 2015-07-20 1 21
Drawings 2015-07-20 38 969
Claims 2015-07-20 7 266
Representative drawing 2015-09-20 1 14
Description 2019-12-19 199 9,509
Claims 2019-12-19 2 60
Claims 2020-11-11 3 101
Claims 2021-07-14 3 101
Representative drawing 2022-05-23 1 14
Confirmation of electronic submission 2024-07-23 2 65
Acknowledgement of Request for Examination 2015-09-09 1 176
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Courtesy - Certificate of registration (related document(s)) 2016-08-09 1 104
Acknowledgement of Request for Examination 2018-12-19 1 189
Commissioner's Notice - Application Found Allowable 2021-10-21 1 572
Electronic Grant Certificate 2022-06-20 1 2,528
New application 2015-07-20 7 243
Correspondence 2015-07-26 1 29
Response to section 37 2015-08-13 73 3,821
Correspondence 2015-08-13 9 329
Courtesy - Office Letter 2015-12-30 1 32
Correspondence 2016-07-18 30 1,186
Correspondence 2016-07-18 30 1,186
Courtesy - Office Letter 2016-08-09 1 32
Modification to the applicant/inventor 2017-05-16 3 80
Courtesy - Filing Certificate for a divisional patent application 2017-06-22 2 111
Examiner Requisition 2019-06-25 5 285
Amendment / response to report 2019-12-19 252 11,443
Examiner requisition 2020-07-12 5 309
Amendment / response to report 2020-11-11 15 521
Interview Record 2021-06-22 1 17
Amendment / response to report 2021-07-14 9 259
Final fee 2022-02-15 4 131
Courtesy - Letter of Remission 2022-10-20 2 365
Courtesy - Letter of Remission 2022-10-20 2 365